22.07.2013 Views

The Australian Immunisation Handbook 10th Edition 2013

The Australian Immunisation Handbook 10th Edition 2013

The Australian Immunisation Handbook 10th Edition 2013

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>The</strong> <strong>Australian</strong><br />

<strong>Immunisation</strong> <strong>Handbook</strong><br />

<strong>10th</strong> <strong>Edition</strong> <strong>2013</strong><br />

i MMUNi SATi ON


Copyright<br />

<strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition <strong>2013</strong><br />

ISBN: 978-1-74241-861-2<br />

Online ISBN: 978-1-74241-862-9<br />

Publications approval number: D0903<br />

Copyright statements:<br />

Paper-based publications<br />

© Commonwealth of Australia <strong>2013</strong><br />

This work is copyright. You may reproduce the whole or part of this work in unaltered form<br />

for your own personal use or, if you are part of an organisation, for internal use within your<br />

organisation, but only if you or your organisation do not use the reproduction for any commercial<br />

purpose and retain this copyright notice and all disclaimer notices as part of that reproduction.<br />

Apart from rights to use as permitted by the Copyright Act 1968 or allowed by this copyright<br />

notice, all other rights are reserved and you are not allowed to reproduce the whole or any part<br />

of this work in any way (electronic or otherwise) without first being given the specific written<br />

permission from the Commonwealth to do so. Requests and inquiries concerning reproduction<br />

and rights are to be sent to the Online, Services and External Relations Branch, Department of<br />

Health and Ageing, GPO Box 9848, Canberra ACT 2601, or via e-mail to copyright@health.gov.au.<br />

Internet sites<br />

© Commonwealth of Australia <strong>2013</strong><br />

This work is copyright. You may download, display, print and reproduce the whole or<br />

part of this work in unaltered form for your own personal use or, if you are part of an<br />

organisation, for internal use within your organisation, but only if you or your organisation<br />

do not use the reproduction for any commercial purpose and retain this copyright notice<br />

and all disclaimer notices as part of that reproduction. Apart from rights to use as permitted<br />

by the Copyright Act 1968 or allowed by this copyright notice, all other rights are reserved<br />

and you are not allowed to reproduce the whole or any part of this work in any way<br />

(electronic or otherwise) without first being given the specific written permission from the<br />

Commonwealth to do so. Requests and inquiries concerning reproduction and rights are<br />

to be sent to the Online, Services and External Relations Branch, Department of Health and<br />

Ageing, GPO Box 9848, Canberra ACT 2601, or via e-mail to copyright@health.gov.au.<br />

<strong>The</strong>se guidelines were approved by the Chief Executive Officer of the National Health and<br />

Medical Research Council (NHMRC) on 25/01/<strong>2013</strong>, under Section 14A of the National Health and<br />

Medical Research Council Act 1992. In approving these guidelines the NHMRC considers that they<br />

meet the NHMRC standard for clinical practice guidelines. This approval is valid for a period of<br />

five years.<br />

NHMRC is satisfied that they are based on the systematic identification and synthesis of the<br />

best available scientific evidence and make clear recommendations for health professionals<br />

practising in an <strong>Australian</strong> health care setting. <strong>The</strong> NHMRC expects that all guidelines will be<br />

reviewed no less than once every five years.<br />

This publication reflects the views of the authors and not necessarily the views of the<br />

<strong>Australian</strong> Government.<br />

ii <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


FOREWORD<br />

Since 1932, when Government vaccination began for <strong>Australian</strong> children, illness<br />

and death from vaccine-preventable diseases have fallen greatly. Australia still has<br />

one of the world’s most comprehensive publicly funded immunisation programs.<br />

As a result, tetanus, diphtheria, Haemophilus influenzae type b, poliomyelitis,<br />

congenital rubella and newly acquired hepatitis B are no longer seen or are<br />

extremely rare.<br />

<strong>Immunisation</strong> is still the safest and most effective way to protect <strong>Australian</strong>s from<br />

vaccine-preventable disease. <strong>The</strong> Government is working towards increasing child<br />

immunisation rates over time by giving parents stronger incentives to have their<br />

children fully immunised.<br />

<strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong>, approved by the National Health and<br />

Medical Research Council (NHMRC), includes clinical information for <strong>Australian</strong><br />

immunisation providers on the safest and most effective use of vaccines, new<br />

vaccines and vaccine-preventable diseases in Australia. It is also a valuable tool to<br />

help immunisation providers explain the benefits of immunisation to their patients.<br />

I’m confident that with your ongoing commitment, immunisation coverage rates<br />

will keep on improving. We need immunisation providers to take every available<br />

opportunity to appropriately vaccinate children and adults. We also need herd<br />

immunity to keep on growing, so that the risk of exposure to vaccine-preventable<br />

diseases such as pertussis and measles is minimised as far as possible.<br />

This <strong>Handbook</strong> includes information on changes to the National <strong>Immunisation</strong><br />

Program. This includes, for example, the recent extension of the<br />

Human Papillomavirus (HPV) Vaccination Program to include males aged<br />

12–13 years. Already the HPV vaccine has had an impact, significantly reducing the<br />

number of lesions that lead to cervical cancer amongst women in the vaccinated<br />

age group. Providing the HPV vaccine to boys will protect them and increase the<br />

effectiveness of the vaccination program for girls.<br />

By building on Australia’s world-class immunisation program, we are stopping<br />

vaccine-preventable diseases and that makes a difference to the quality of life for<br />

<strong>Australian</strong> families.<br />

Finally, I would like to thank the Chair, Professor Terry Nolan, and members<br />

of the <strong>Australian</strong> Technical Advisory Group on <strong>Immunisation</strong>, its working<br />

parties, technical writers and advisors for their work in producing this excellent<br />

resource. It will be a great help to everyone involved in supporting and delivering<br />

immunisation services in Australia.<br />

<strong>The</strong> Hon Tanya Plibersek MP<br />

Minister for Health<br />

iii


iv <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


TABLE OF CONTENTS<br />

PART 1 INTRODUCTION TO THE AUSTRALIAN IMMUNISATION HANDBOOK 1<br />

1.1 Background 1<br />

1.2 Development of the <strong>10th</strong> edition of the <strong>Handbook</strong> 3<br />

1.3 How to use the <strong>10th</strong> edition <strong>Handbook</strong> 5<br />

1.4 What’s new 7<br />

1.5 Fundamentals of immunisation 18<br />

PART 2 VACCINATION PROCEDURES 24<br />

2.1 Pre-vaccination 24<br />

2.2 Administration of vaccines 65<br />

2.3 Post-vaccination 85<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 104<br />

3.1 Vaccination for Aboriginal and Torres Strait Islander people 104<br />

3.2 Vaccination for international travel 113<br />

3.3 Groups with special vaccination requirements 130<br />

PART 4 VACCINE-PREVENTABLE DISEASES 176<br />

4.1 Cholera 176<br />

4.2 Diphtheria 182<br />

4.3 Haemophilus influenzae type b 191<br />

4.4 Hepatitis A 198<br />

4.5 Hepatitis B 208<br />

4.6 Human papillomavirus 231<br />

4.7 Influenza 243<br />

4.8 Japanese encephalitis 259<br />

4.9 Measles 267<br />

4.10 Meningococcal disease 283<br />

4.11 Mumps 295<br />

4.12 Pertussis 302<br />

4.13 Pneumococcal disease 317<br />

4.14 Poliomyelitis 338<br />

4.15 Q fever 345<br />

4.16 Rabies and other lyssaviruses (including <strong>Australian</strong> bat lyssavirus) 353<br />

v


4.17 Rotavirus 372<br />

4.18 Rubella 384<br />

4.19 Tetanus 397<br />

4.20 Tuberculosis 408<br />

4.21 Typhoid 416<br />

4.22 Varicella 423<br />

4.23 Yellow fever 439<br />

4.24 Zoster (herpes zoster) 446<br />

PART 5 PASSIVE IMMUNISATION 456<br />

5.1 Passive immunisation using immunoglobulin preparations 456<br />

APPENDICES 465<br />

APPENDIX 1: Contact details for <strong>Australian</strong>, state and territory government<br />

health authorities and communicable disease control 465<br />

APPENDIX 2: Literature search strategy for the <strong>10th</strong> edition of the <strong>Handbook</strong> 467<br />

APPENDIX 3: Components of vaccines used in the National<br />

<strong>Immunisation</strong> Program 469<br />

APPENDIX 4: Commonly asked questions about vaccination 473<br />

A.4.1 General questions 473<br />

A4.2 Questions about contraindications and precautions 477<br />

A4.3 Questions about vaccine safety 481<br />

A4.4 Questions about vaccine content 484<br />

A4.5 Questions about the need for immunisation 487<br />

A4.6 Further information about vaccination 488<br />

APPENDIX 5: Glossary of technical terms 489<br />

APPENDIX 6: Commonly used abbreviations 495<br />

APPENDIX 7: Overview of vaccine availability in Australia 498<br />

INDEX 500<br />

vi <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


LIST OF TABLES<br />

Table 2.1.1: Pre-vaccination screening checklist 30<br />

Table 2.1.2: Responses to relevant conditions or circumstances identified<br />

through the pre-vaccination screening checklist 31<br />

Table 2.1.3: Live attenuated parenteral and oral vaccines 37<br />

Table 2.1.4: False contraindications to vaccination 38<br />

Table 2.1.5: Minimum acceptable age for the 1st dose of scheduled vaccines<br />

in infants in special circumstances 46<br />

Table 2.1.6: Number of vaccine doses that should have been administered<br />

by the current age of the child 49<br />

Table 2.1.7: Minimum acceptable dose intervals for children


Table 3.1.1: Additional vaccines recommended for Indigenous persons,<br />

due to their higher risk of disease 105<br />

Table 3.2.1: Dose and routes of administration of commonly used vaccines<br />

in adult travellers 123<br />

Table 3.2.2: Recommended lower age limits of travel vaccines for children 127<br />

Table 3.3.1: Recommendations for vaccination in pregnancy 135<br />

Table 3.3.2: Recommendations for vaccinations for solid organ transplant<br />

(SOT) recipients 151<br />

Table 3.3.3: Recommendations for revaccination following haematopoietic<br />

stem cell transplant (HSCT) in children and adults, irrespective<br />

of previous immunisation history 156<br />

Table 3.3.4: Categories of immunocompromise in HIV-infected persons, based<br />

on age-specific CD4 + counts and percentage of total lymphocytes 158<br />

Table 3.3.5: Recommendations for vaccination in persons with functional or<br />

anatomical asplenia 163<br />

Table 3.3.6: Recommended intervals between either immunoglobulins or<br />

blood products and measles-mumps-rubella (MMR), measlesmumps-rubella-varicella<br />

(MMRV) or varicella vaccination 167<br />

Table 3.3.7: Recommended vaccinations for persons at increased risk of<br />

certain occupationally acquired vaccine-preventable diseases 170<br />

Table 4.4.1: Recommended doses and schedules for use of inactivated<br />

hepatitis A and hepatitis A combination vaccines 202<br />

Table 4.5.1: Recommended schedules for use of monovalent hepatitis B and<br />

hepatitis B combination vaccines 214<br />

Table 4.5.2: Accelerated hepatitis B vaccination schedules (for persons<br />

with imminent risk of exposure) 216<br />

Table 4.5.3: Post-exposure prophylaxis for non-immune persons exposed<br />

to a HBsAg-positive source 229<br />

Table 4.7.1: Recommended doses of influenza vaccine 251<br />

Table 4.9.1: Recommendations for measles vaccination with (a) measlesmumps-rubella<br />

(MMR) (currently available), and (b) once<br />

measles-mumps-rubella-varicella (MMRV) vaccines are available<br />

from July <strong>2013</strong> 273<br />

Table 4.9.2: Post-exposure prophylaxis ≤72 hours since exposed to measles<br />

for non-immune individuals (adapted from Measles: national<br />

guidelines for public health units) 281<br />

Table 4.13.1: Recommendations for pneumococcal vaccination for children<br />

aged


List 4.13.1: Conditions associated with an increased risk of invasive<br />

pneumococcal disease (IPD) in children and adults, by severity<br />

of risk 326<br />

Table 4.13.2: Recommendations for pneumococcal vaccination for children<br />

aged 2–5 years with a medical condition(s) associated with an<br />

increased risk of invasive pneumococcal disease (IPD) 328<br />

Table 4.13.3: Recommendations for pneumococcal vaccination using 23vPPV<br />

for adults who do not have a condition(s) associated with an<br />

increased risk of invasive pneumococcal disease (IPD) 332<br />

Table 4.15.1: Interpretation and action for serological and skin test results<br />

(with modifications from A guide to Q fever and Q fever vaccination<br />

(CSL Biotherapies, 2009)) 350<br />

Table 4.16.1: Lyssavirus exposure categories 360<br />

Table 4.16.2: Post-exposure prophylaxis commenced overseas and<br />

recommended completion in Australia 365<br />

Table 4.17.1: Upper age limits for dosing of oral rotavirus vaccines 378<br />

Table 4.19.1: Guide to tetanus prophylaxis in wound management 404<br />

Table 4.22.1: Recommendations for varicella vaccination with (a) monovalent<br />

varicella vaccine (VV) (currently available), and (b) once measlesmumps-rubella-varicella<br />

(MMRV) vaccines are available from<br />

July <strong>2013</strong> 428<br />

Table 4.22.2: Zoster immunoglobulin-VF (ZIG) dose based on weight 436<br />

Table A2.1: Electronic databases searched for the <strong>10th</strong> edition 467<br />

Table A3.1: Components of vaccines used in the National<br />

<strong>Immunisation</strong> Program 469<br />

Table A7.1: Key dates when vaccines first came into widespread<br />

use in Australia 498<br />

ix


LIST OF FIGURES<br />

Figure 2.1.1: Catch-up worksheet for children


PREFACE<br />

<strong>The</strong> <strong>10th</strong> edition of <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> was prepared by<br />

the <strong>Australian</strong> Technical Advisory Group on <strong>Immunisation</strong> of the <strong>Australian</strong><br />

Government Department of Health and Ageing.<br />

Members of the <strong>Australian</strong> Technical Advisory Group on<br />

<strong>Immunisation</strong><br />

Chair<br />

Professor Terry Nolan, Paediatrician; Epidemiologist and Head, School of<br />

Population Health, <strong>The</strong> University of Melbourne, Victoria<br />

Deputy Chair<br />

Associate Professor Peter Richmond, School of Paediatrics and Child Health,<br />

<strong>The</strong> University of Western Australia; General Paediatrician and Paediatric<br />

Immunologist, Princess Margaret Hospital for Children, Western Australia<br />

Voting and ex-officio members<br />

Associate Professor Ross Andrews, Menzies School of Health Research,<br />

Northern Territory<br />

Associate Professor Christopher Blyth, School of Paediatrics and Child Health,<br />

Faculty of Medicine, Dentistry and Health Sciences, <strong>The</strong> University of Western<br />

Australia, Princess Margaret Hospital, Western Australia (member from July 2012)<br />

Ms Sue Campbell-Lloyd, Manager, <strong>Immunisation</strong> Unit, AIDS/Infectious Diseases<br />

Branch, NSW Health, New South Wales (member until June 2012)<br />

Dr Grahame Dickson, Medical Officer, Drug Safety and Evaluation Branch,<br />

<strong>The</strong>rapeutic Goods Administration, <strong>Australian</strong> Government Department of<br />

Health and Ageing, <strong>Australian</strong> Capital Territory (member until August 2012)<br />

Dr Nicole Gilroy, Infectious Diseases consultant, St Vincent’s Hospital, Sydney;<br />

Blood Marrow Transplant (BMT) Network, New South Wales<br />

Ms Madeline Hall, Public Health Nurse, Queensland Health, Queensland<br />

(member from July 2012)<br />

Clinical Professor David Isaacs, Senior Staff Specialist, Department of Infectious<br />

Diseases and Microbiology, <strong>The</strong> Children’s Hospital at Westmead and Paediatric<br />

Infectious Diseases, <strong>The</strong> University of Sydney, New South Wales<br />

Dr Rosemary Lester, Chief Health Officer, Department of Health, Victoria<br />

(member until December 2011)<br />

xi


Dr Ting Lu, Medical Officer, Office of Medicines Authorisation, Market<br />

Authorisation Group, <strong>The</strong>rapeutic Goods Administration, <strong>Australian</strong><br />

Government Department of Health and Ageing, <strong>Australian</strong> Capital Territory<br />

(member from September 2012)<br />

Professor Peter McIntyre, Professor of Paediatrics and Preventive Medicine,<br />

<strong>The</strong> University of Sydney; Director, National Centre for <strong>Immunisation</strong> Research<br />

and Surveillance of Vaccine Preventable Diseases, <strong>The</strong> Children’s Hospital at<br />

Westmead, New South Wales<br />

Associate Professor Jodie McVernon, School of Population Health, <strong>The</strong> University<br />

of Melbourne, Victoria (member from January 2012)<br />

Dr Joanne Molloy, General Practitioner, GP Association of Geelong, Victoria<br />

Ms Stephanie Newell, Consumer representative (member until June 2012)<br />

Associate Professor Michael Nissen, Director of Infectious Diseases and<br />

Clinical Microbiologist, Unit Head of Queensland Paediatric Infectious Disease<br />

Laboratory; Associate Professor in Biomolecular, Biomedical Science and Health,<br />

Royal Children’s Hospital, Queensland (member until June 2012)<br />

Dr Rod Pearce, General Practitioner, Medical Officer of Health, Eastern Health<br />

Authority, Adelaide; GP <strong>Immunisation</strong> Advisor, Adelaide Central and Eastern<br />

Division of General Practice, South Australia (member until June 2012)<br />

Ms Karen Peterson, <strong>Immunisation</strong> Manager, Queensland Health, Queensland<br />

(member from July 2012)<br />

Ms Debra Petrys, Consumer representative (member from July 2012)<br />

Ms Helen Pitcher, Public Health Nurse, Department of Human Services, Victoria<br />

(member until June 2012)<br />

Ms Julianne Quaine, Assistant Secretary, Health Protection Programs Branch,<br />

Office of Health Protection, <strong>Australian</strong> Government Department of Health and<br />

Ageing, <strong>Australian</strong> Capital Territory<br />

Dr Greg Rowles, General Practitioner, Riddells Creek, Victoria (member from<br />

July 2012)<br />

Dr Christine Selvey, Communicable Diseases Network Australia, Queensland<br />

Health, Queensland (member from January 2012)<br />

Professor Steven Wesselingh, Executive Director, South <strong>Australian</strong> Health and<br />

Medical Research Institute, South Australia (member from July 2012)<br />

Secretary<br />

Ms Monica Johns, Director, <strong>Immunisation</strong> Policy Section, <strong>Immunisation</strong> Branch,<br />

Office of Health Protection, <strong>Australian</strong> Government Department of Health and<br />

Ageing, <strong>Australian</strong> Capital Territory<br />

xii <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Secretariat support, <strong>Australian</strong> Technical Advisory Group on<br />

<strong>Immunisation</strong><br />

Ms Sandy Anderson; Ms Jessica Hutchison<br />

Senior technical editor<br />

Associate Professor Kristine Macartney, Deputy Director Government Programs,<br />

National Centre for <strong>Immunisation</strong> Research and Surveillance of Vaccine<br />

Preventable Diseases, <strong>The</strong> Children’s Hospital at Westmead; Discipline of<br />

Paediatrics and Child Health, <strong>The</strong> University of Sydney<br />

Technical editor<br />

Dr Jane Jelfs, Manager Policy Support, National Centre for <strong>Immunisation</strong><br />

Research and Surveillance of Vaccine Preventable Diseases<br />

Assistant technical editor<br />

Dr Melina Georgousakis, Research Officer, National Centre for <strong>Immunisation</strong><br />

Research and Surveillance of Vaccine Preventable Diseases<br />

Senior technical writer<br />

Dr Clayton Chiu, Public Health Physician, National Centre for <strong>Immunisation</strong><br />

Research and Surveillance of Vaccine Preventable Diseases<br />

Technical writers<br />

Mr Brett Archer; Ms Kathryn Cannings; Dr Bradley Christian; Dr Nigel<br />

Crawford; Dr Aditi Dey; Dr Anita Heywood; Dr Sanjay Jayasinghe; Dr Robert<br />

Menzies; Dr Helen Quinn; Dr Tom Snelling; Ms Kirsten Ward; Dr Nicholas Wood<br />

Editorial support<br />

Ms Donna Armstrong, Editing and Publications Officer, National Centre for<br />

<strong>Immunisation</strong> Research and Surveillance of Vaccine Preventable Diseases<br />

Library support<br />

Ms Catherine King, Information Manager, National Centre for <strong>Immunisation</strong><br />

Research and Surveillance of Vaccine Preventable Diseases<br />

Mr Edward Jacyna, Assistant Librarian, National Centre for <strong>Immunisation</strong><br />

Research and Surveillance of Vaccine Preventable Diseases<br />

Administration support<br />

Ms Lyn Benfield, Senior Administration Project Officer, National Centre for<br />

<strong>Immunisation</strong> Research and Surveillance of Vaccine Preventable Diseases<br />

xiii


Acknowledgments<br />

Dr Frank Beard<br />

Professor Julie Bines<br />

Dr Julia Brotherton<br />

Associate Professor Tony Brown<br />

Professor Margaret Burgess<br />

Dr Dave Burgner<br />

Dr Jim Buttery<br />

Professor Jonathan Carapetis<br />

Dr Ben Cowie<br />

Dr Andrew Daley<br />

Professor Basil Donovan<br />

Dr Mark Douglas<br />

Professor David Durrheim<br />

Professor Dominic Dwyer<br />

Professor Joan Faoagali<br />

Dr Tony Gherardin<br />

Dr Heather Gidding<br />

Professor Lyn Gilbert<br />

Dr Robert Hall<br />

Dr Alan Hampson<br />

Dr Penny Hutchinson<br />

Dr Heath Kelly<br />

Professor Michael Kidd<br />

Dr Ann Koehler<br />

xiv <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Dr Vicki Krause<br />

Associate Professor Stephen Lambert<br />

Associate Professor Amanda Leach<br />

Professor Raina MacIntyre<br />

Professor John Mackenzie<br />

Associate Professor Guy Marks<br />

Dr Jeremy McAnulty<br />

Dr Brad McCall<br />

Dr Elizabeth McCarthy<br />

Dr Neil Parker<br />

Professor Bill Rawlinson<br />

Associate Professor Tilman Ruff<br />

Dr Rosalie Schultz<br />

Dr Vicky Sheppeard<br />

Associate Professor Vitali Sintchenko<br />

Associate Professor Monica Slavin<br />

Associate Professor David Smith<br />

Dr Bruce Thorley<br />

Dr Joe Torressi<br />

Dr Siranda Torvaldsen<br />

Ms Maureen Watson<br />

Dr Rosalind Webby<br />

Dr Melanie Wong<br />

Professor Nick Zwar


PART 1 INTRODUCTION TO THE AUSTRALIAN<br />

IMMUNISATION HANDBOOK<br />

1.1 BACKGROUND<br />

For more than 200 years, since Edward Jenner first demonstrated that<br />

vaccination offered protection against smallpox, the use of vaccines has<br />

continued to reduce the burden of many infectious diseases. Vaccination has<br />

been demonstrated to be one of the most effective and cost-effective public<br />

health interventions. Worldwide, it has been estimated that immunisation<br />

programs prevent approximately 2.5 million deaths each year. 1 <strong>The</strong> global<br />

eradication of smallpox in 1997, near elimination of poliomyelitis and global<br />

reduction in other vaccine-preventable diseases, are model examples of disease<br />

control through immunisation.<br />

Vaccination not only protects individuals, but also protects others in the<br />

community by increasing the overall level of immunity in the population<br />

and thus minimising the spread of infection. This concept is known as ‘herd<br />

immunity’. It is vital that healthcare professionals take every available<br />

opportunity to vaccinate children and adults. Australia has one of the most<br />

comprehensive publicly funded immunisation programs in the world. As a result<br />

of successful vaccination programs in Australia, many diseases, for example,<br />

tetanus, diphtheria, Haemophilus influenzae type b and poliomyelitis, do not occur<br />

now or are extremely rare in Australia. 2<br />

<strong>The</strong> purpose of <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> is to provide clinical<br />

guidelines for health professionals on the safest and most effective use of vaccines<br />

in their practice. <strong>The</strong>se recommendations are developed by the <strong>Australian</strong><br />

Technical Advisory Group on <strong>Immunisation</strong> (ATAGI) and were considered<br />

for approval by the National Health and Medical Research Council (NHMRC)<br />

(under section 14A of the NHMRC Act 1992).<br />

<strong>The</strong> <strong>Handbook</strong> provides guidance based on the best scientific evidence available<br />

at the time of publication from published and unpublished literature. Further<br />

details regarding the <strong>Handbook</strong> revision procedures are described below in 1.2<br />

Development of the <strong>10th</strong> edition of the <strong>Handbook</strong>. <strong>The</strong> reference lists for all chapters<br />

are included in the electronic version of the <strong>Handbook</strong>, which is available via the<br />

Immunise Australia website (www.immunise.health.gov.au).<br />

<strong>The</strong> information contained within this <strong>Handbook</strong> was correct as at October 2012.<br />

However, the content of the <strong>Handbook</strong> is reviewed regularly. <strong>The</strong> <strong>10th</strong> edition<br />

of <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> will remain current unless amended<br />

electronically via the Immunise Australia website or until the 11th edition of the<br />

<strong>Handbook</strong> is published.<br />

PART 1 INTRODUCTION TO THE AUSTRALIAN IMMUNISATION HANDBOOK 1<br />

1.1 BACKGROUND


Information is provided in the <strong>Handbook</strong> for all vaccines that are available in<br />

Australia at or near the time of publication. <strong>The</strong>se include many vaccines that are<br />

funded under the National <strong>Immunisation</strong> Program (NIP). A copy of the current<br />

NIP schedule is provided with the hard copy of the <strong>Handbook</strong>. However, the<br />

NIP schedule may also be updated regularly; immunisation service providers<br />

should consult the Immunise Australia website (www.immunise.health.gov.<br />

au) for changes. A number of vaccines included in this <strong>Handbook</strong> are not part<br />

of the routine immunisation schedule; these vaccines may be given to, for<br />

example, persons travelling overseas, persons with a medical condition placing<br />

them at increased risk of contracting a vaccine-preventable disease, or those at<br />

occupational risk of disease.<br />

Electronic updates to the <strong>10th</strong> edition of<br />

<strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> will be available at:<br />

www.immunise.health.gov.au<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

2 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


1.2 DEVELOPMENT OF THE 10TH EDITION OF THE HANDBOOK<br />

<strong>The</strong> <strong>10th</strong> edition of the <strong>Handbook</strong> has been developed by the <strong>Australian</strong> Technical<br />

Advisory Group on <strong>Immunisation</strong> (ATAGI), which provides advice to the Federal<br />

Minister for Health on the Immunise Australia Program and other vaccinerelated<br />

issues. In addition to technical experts from many fields, the ATAGI’s<br />

membership includes a consumer representative and general practitioners. Staff<br />

of the National Centre for <strong>Immunisation</strong> Research and Surveillance of Vaccine<br />

Preventable Diseases (NCIRS) provided the technical support to the ATAGI to<br />

develop the <strong>Handbook</strong>.<br />

It is important to note that recommendations contained within the <strong>Handbook</strong><br />

do not formally address the cost-effectiveness of different vaccines or different<br />

vaccine schedules. Since January 2006, the cost-effectiveness of vaccines<br />

is assessed by the Pharmaceutical Benefits Advisory Committee (PBAC),<br />

which advises government on the funding of vaccines under the National<br />

<strong>Immunisation</strong> Program and/or Pharmaceutical Benefits Scheme (PBS). Most,<br />

but not all, of the recommendations made within the <strong>Handbook</strong> will be funded<br />

under the NIP, PBS or via other means, such as through special schemes and<br />

state- or territory-based programs.<br />

1.2.1 Process of developing <strong>Handbook</strong> recommendations<br />

<strong>The</strong> <strong>Handbook</strong> is designed as a general guide to inform clinicians on the safest and<br />

most effective vaccination strategies, using the highest quality evidence available.<br />

In the absence of high-quality evidence, such as well-conducted randomised<br />

controlled trials and meta-analyses, the ATAGI based its recommendations on<br />

less rigorous studies, such as uncontrolled clinical trials, case-series and/or other<br />

observational studies. Where clinical guidelines were available on specific topics,<br />

these were also consulted to help frame recommendations, if relevant, in the<br />

<strong>Australian</strong> setting. Further details on literature search strategies utilised for the<br />

production of this edition can be found in Appendix 2. <strong>The</strong> ATAGI also consulted<br />

immunisation handbooks produced by comparable countries. When published<br />

sources were inadequate, recommendations were based on expert opinion.<br />

However, limitations and challenges to developing recommendations continue to<br />

exist when there are unaddressed scientific questions, complex medical practice<br />

issues and continuous new information, as well as differences in expert opinion.<br />

Despite these limitations, the ATAGI has sought to provide clear and relevant<br />

recommendations wherever possible.<br />

<strong>The</strong> 1st edition of <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> was published in 1975.<br />

Due to its longevity, scope and complexity, the <strong>Handbook</strong> does differ from other<br />

NHMRC guidelines. As such, recommendations contained in the <strong>Handbook</strong> do<br />

not contain formal levels or grades of evidence or evidence tables. <strong>The</strong> grades of<br />

evidence assigned to recommendations for the three newly vaccine-preventable<br />

diseases (human papillomavirus [HPV], rotavirus and herpes zoster) that were<br />

included in the 9th edition <strong>Handbook</strong> (and its electronic amendments) have now<br />

PART 1 INTRODUCTION TO THE AUSTRALIAN IMMUNISATION HANDBOOK 3<br />

1.2 DEVELOPMENT OF THE 10TH OF EDITION THE 10TH OF THE HANDBOOK<br />

EDITION OF THE HANDBOOK


een removed and replaced with an evidence statement for consistency with<br />

other chapters in the <strong>10th</strong> edition of the <strong>Handbook</strong>. <strong>The</strong> ATAGI has developed new<br />

recommendations for the <strong>Handbook</strong> after considering clinical questions, reviewing<br />

available evidence, as described above, and through extensive consultation with<br />

other experts (described below). Evidence statements for recommendations, with<br />

cross reference to other relevant sections of the <strong>Handbook</strong>, have been included<br />

wherever possible.<br />

1.2.2 Consultation and input into the draft <strong>10th</strong> edition <strong>Handbook</strong><br />

Prior to completion of the draft <strong>10th</strong> edition <strong>Handbook</strong>, the ATAGI sought expert<br />

review of individual chapters by leading <strong>Australian</strong> experts. <strong>The</strong>se reviewers,<br />

who are acknowledged in the front of this <strong>Handbook</strong>, provided input to further<br />

refine each chapter. <strong>The</strong> ATAGI also, where relevant, consulted members of<br />

each of its current disease-based Working Parties. Other peak advisory groups,<br />

in particular the National <strong>Immunisation</strong> Committee, were also consulted and<br />

provided valuable input into the development of this revised edition. <strong>The</strong> draft<br />

<strong>10th</strong> edition of the <strong>Handbook</strong> was available for public consultation over a 4-week<br />

period during July–August 2012. <strong>The</strong> ATAGI reviewed all public comments<br />

received and, where necessary, incorporated these as changes to the <strong>Handbook</strong>.<br />

<strong>The</strong> NHMRC was also consulted throughout the development of the <strong>10th</strong><br />

edition of the <strong>Handbook</strong>, prior to the <strong>Handbook</strong> being proposed for submission to<br />

the NHMRC for consideration of approval (under section 14A of the NHMRC<br />

Act 1992).<br />

1.2.3 Implementation of recommendations in the <strong>10th</strong> edition<br />

<strong>Handbook</strong><br />

<strong>The</strong> <strong>10th</strong> edition of the <strong>Handbook</strong> is disseminated directly to all registered<br />

medical practitioners in Australia. Additional hard copies are distributed to<br />

other immunisation service providers via their state or territory health authority.<br />

An electronic version of the <strong>Handbook</strong> is freely accessible on the Immunise<br />

Australia Program website (www.immunise.health.gov.au). Implementation of<br />

the recommendations as stated in the <strong>Handbook</strong> is undertaken by immunisation<br />

service providers in conjunction with their state or territory health authority and<br />

the Immunise Australia Program of the <strong>Australian</strong> Government Department of<br />

Health and Ageing.<br />

4 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


1.3 HOW TO USE THE 10TH EDITION HANDBOOK<br />

<strong>The</strong> following information provides an overview of how information is presented<br />

in this <strong>10th</strong> edition of <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong>, to guide the reader<br />

in how to best use this resource.<br />

<strong>The</strong> <strong>Handbook</strong> is divided into five major parts. Part 1 contains information on<br />

the development process for the <strong>Handbook</strong>, a summary of changes from the 9th<br />

edition <strong>Handbook</strong>, and an overview of the general principles of immunisation,<br />

including both active and passive immunisation, vaccine efficacy and vaccine<br />

safety. Part 2 of the <strong>Handbook</strong> is divided into three chapters, which describe<br />

the processes and procedures involved around a vaccination encounter: prevaccination<br />

requirements; a detailed discussion of the administration of vaccines;<br />

and information on post-vaccination considerations. In Part 3 of the <strong>Handbook</strong>,<br />

there are three chapters, each dedicated to vaccinations for different special risk<br />

groups, including Aboriginal and Torres Strait Islander people, international<br />

travellers, and other special risk categories (e.g. immunocompromised persons,<br />

pregnant women and others).<br />

Part 4 of the <strong>Handbook</strong> contains 24 chapters, each dealing with an individual<br />

disease for which a vaccine, or vaccines, are currently available in Australia.<br />

<strong>The</strong>se chapters all follow the same format and are divided into various sections<br />

describing:<br />

• the biology, clinical features and epidemiology of the disease<br />

• information on vaccines, with those available, or soon to be available, in<br />

Australia highlighted in a shaded box (for more information see also the<br />

<strong>The</strong>rapeutic Goods Administration, www.tga.gov.au, for the latest vaccine<br />

product information document provided by the manufacturer and<br />

1.5.3 Active immunisation)<br />

• recommendations for vaccine use<br />

• use in pregnancy and breastfeeding<br />

• contraindications, precautions and advice on adverse events following<br />

immunisation specific to the vaccine(s) (for more information see<br />

1.5.5 Vaccine safety and adverse events following immunisation)<br />

• information on the public health management of each disease; this is<br />

only given in detail where there are specific additional recommendations<br />

for vaccine use in the context of disease control and/or post-exposure<br />

prophylaxis<br />

• variations from product information.<br />

PART 1 INTRODUCTION TO THE AUSTRALIAN IMMUNISATION HANDBOOK 5<br />

1.3 HOW TO USE THE<br />

10TH EDITION HANDBOOK


Variations from product information<br />

In some instances, the ATAGI recommendations in the <strong>Handbook</strong><br />

may differ from information provided by the manufacturer in the<br />

vaccine product information document (PI); these differences may be<br />

recommendations that are in addition to or instead of those listed in the<br />

PI. Where indicated, variations from the PI are detailed in each relevant<br />

vaccine chapter under the heading ‘Variations from product information’.<br />

Where a variation exists, the ATAGI recommendation should be<br />

considered best practice.<br />

<strong>The</strong> reader is encouraged to seek additional information on communicable<br />

disease surveillance, prevention and control, including published guidelines,<br />

from the Communicable Diseases Network Australia (CDNA) via the <strong>Australian</strong><br />

Government CDNA website (www.health.gov.au/cdnasongs).<br />

Part 5 of the <strong>Handbook</strong> pertains to passive immunisation using immunoglobulin<br />

preparations and provides an overview of the available products and their<br />

intended use.<br />

<strong>The</strong>re are a number of appendices, including contact details for state and<br />

territory health departments (Appendix 1); the literature search strategies<br />

used for the <strong>10th</strong> edition of the <strong>Handbook</strong> (Appendix 2); components used in<br />

the vaccines available via the National <strong>Immunisation</strong> Program (Appendix 3);<br />

some commonly asked questions (Appendix 4); a glossary of terms used in the<br />

<strong>Handbook</strong> (Appendix 5); abbreviations used in the <strong>Handbook</strong> (Appendix 6); and a<br />

list of dates when various vaccines became available in Australia (Appendix 7).<br />

6 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


1.4 WHAT’S NEW<br />

All chapters have been updated and revised, where necessary, from the 9th<br />

edition. <strong>The</strong> <strong>10th</strong> edition introduces new vaccines, changes to the schedules and<br />

recommendations, and changes to the presentation of the <strong>Handbook</strong>.<br />

1.4.1 New chapters and chapters that no longer appear in<br />

the <strong>Handbook</strong><br />

• Layout of the <strong>Handbook</strong> differs from the previous edition. <strong>The</strong> <strong>Handbook</strong><br />

is now in 5 parts: Part 1 Introduction to <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong><br />

<strong>Handbook</strong>; Part 2 Vaccination procedures; Part 3 Vaccination for special risk<br />

groups; Part 4 Vaccine-preventable diseases; and Part 5 Passive immunisation.<br />

• Part 1 now includes the development process for the <strong>Handbook</strong><br />

(previously in Appendix 2) and information on the fundamentals of<br />

immunisation, including passive and active immunisation, vaccine<br />

efficacy and vaccine safety.<br />

• <strong>The</strong> new Part 5 Passive immunisation contains information previously<br />

contained in Chapter 3.8 of the 9th edition, Immunoglobulin preparations.<br />

• <strong>The</strong> chapter on <strong>Australian</strong> bat lyssavirus and rabies is now listed under Rabies<br />

and other lyssaviruses (including <strong>Australian</strong> bat lyssavirus) in the alphabetical list<br />

of diseases in Part 4.<br />

• <strong>The</strong> chapter on zoster (herpes zoster) is now included as a disease chapter<br />

in Part 4 in this printed version of the <strong>10th</strong> edition. It had previously been<br />

available as an online update to the 9th edition.<br />

• <strong>The</strong> chapter on smallpox has been deleted. For information on smallpox,<br />

please go to the <strong>Australian</strong> Government website www.health.gov.au and<br />

use the index option to select the smallpox fact sheet.<br />

• Some appendices contained in the 9th edition have been deleted:<br />

» Products registered in Australia but not currently available (previously<br />

Appendix 3); information regarding such products can be sought from<br />

the <strong>The</strong>rapeutic Goods Administration (www.tga.gov.au)<br />

» Definitions of adverse events following immunisation (previously Appendix 6);<br />

some common adverse events are now defined in the glossary of technical<br />

terms and an expanded section on adverse events following immunisation<br />

is now provided in Part 2 (2.3.2)<br />

» Summary table – procedures for a vaccination encounter (previously Appendix 10);<br />

this information is now incorporated throughout Part 2.<br />

1.4.2 Changes to the format of disease chapters in Part 4<br />

• Where relevant, information on reconstitution and stability of reconstituted<br />

vaccines has been included in the ‘Transport, storage and handling’ section of<br />

disease chapters.<br />

PART 1 INTRODUCTION TO THE AUSTRALIAN IMMUNISATION HANDBOOK 7<br />

1.4 WHAT'S NEW


• Where relevant, information on co-administration with other vaccines<br />

and interchangeability of vaccines is now included in the ‘Dosage and<br />

administration’ section of disease chapters.<br />

• A ‘Pregnancy and breastfeeding’ section has been added to all disease<br />

chapters.<br />

• Information on the public health management of each disease is only given<br />

in detail where there are specific additional recommendations for vaccine<br />

use in the context of disease control and/or post-exposure prophylaxis.<br />

<strong>The</strong> reader is referred to published guidelines from the Communicable<br />

Diseases Network Australia (CDNA), where available.<br />

• <strong>The</strong> Evidence Grades assigned in the 9th edition to recommendations<br />

contained within three chapters – Human papillomavirus, Rotavirus and Zoster<br />

(online only) – have been removed (see discussion in 1.2 Development of the<br />

<strong>10th</strong> edition of the <strong>Handbook</strong> regarding <strong>Handbook</strong> development).<br />

1.4.3 Overview of major changes to recommendations<br />

<strong>The</strong> following list summarises major changes to recommendations and other<br />

important information that have occurred in each part of the <strong>10th</strong> edition of<br />

the <strong>Handbook</strong>.<br />

Part 2 Vaccination procedures<br />

2.1 Pre-vaccination<br />

• <strong>The</strong> checklist summarising activities required for optimal storage of vaccines<br />

has been deleted; readers are referred to the National vaccine storage guidelines:<br />

Strive for 5.<br />

• <strong>The</strong> table (Table 2.1.5) containing information on the minimum acceptable<br />

age for the 1st vaccine doses in infants now provides advice on action<br />

required in case of early administration.<br />

• Catch-up recommendations and tables are now for children aged


2.2 Administration of vaccines<br />

• Advice is now provided on the use of vaccines in multi-dose vials.<br />

• Advice is provided on what to do if a vaccine is inadvertently administered<br />

via a route (e.g. intramuscular [IM] or subcutaneous [SC]) other than for<br />

which it is recommended.<br />

• Information is now provided on vaccinating children with congenital limb<br />

malformation, children in spica casts, patients undergoing treatment for<br />

breast cancer, and patients with lymphoedema.<br />

• <strong>The</strong> section on administration of multiple vaccine injections at the same visit<br />

now includes advice on the order in which to give sequential vaccines and<br />

advice on simultaneous injections by two providers.<br />

2.3 Post-vaccination<br />

• <strong>The</strong> section on adverse events following immunisation has been enhanced<br />

and expanded, including use of adrenaline autoinjectors for anaphylaxis<br />

treatment and more information on reporting of adverse events following<br />

immunisation.<br />

• Contact details are provided for obtaining HPV vaccination history from<br />

the National HPV Vaccination Program Register (NHVPR, or the ‘HPV<br />

Register’).<br />

• <strong>The</strong> section on documentation of vaccination has been expanded to include<br />

updated details for reporting to the ACIR, and information on the National<br />

HPV Vaccination Program Register and other registers.<br />

Part 3 Vaccination for special risk groups<br />

3.1 Vaccination for Aboriginal and Torres Strait Islander people<br />

• Information on the burden of influenza in Indigenous children, and the<br />

rationale for vaccination of those, especially ≥6 months to


3.2 Vaccination for international travel<br />

• This section has been updated and expanded and information on<br />

recommended vaccines is now divided into routinely recommended vaccines<br />

(that are not specifically related to travelling overseas) and selected vaccines<br />

that are recommended based on travel itinerary, activities and likely risk of<br />

disease exposure.<br />

• Information on more vaccines, including new vaccines, has been added to<br />

the tables outlining the dose and routes of administration (Table 3.2.1) and<br />

recommended lower age limits (Table 3.2.2) for vaccines for travellers.<br />

3.3 Groups with special vaccination requirements<br />

• <strong>The</strong> section on vaccination of persons with a prior adverse event following<br />

immunisation has been expanded. Advice is provided on vaccination of<br />

persons with allergies, including egg allergy.<br />

• <strong>The</strong> section on vaccination of women who are planning pregnancy, pregnant<br />

or breastfeeding, and preterm infants has been updated and expanded.<br />

• <strong>The</strong> table of recommendations for vaccination in pregnancy (Table 3.3.1) has<br />

been updated to include new vaccines.<br />

• dTpa vaccine can be given during the third trimester of pregnancy as an<br />

alternative to post-partum or pre-conception vaccination.<br />

• <strong>The</strong> section on vaccination of immunocompromised persons, including<br />

transplant recipients and oncology patients, has been updated and expanded.<br />

• All immunocompromised persons, irrespective of age, who receive influenza<br />

vaccine for the first time are now recommended to receive 2 vaccine doses, at<br />

least 4 weeks apart, and 1 dose annually thereafter.<br />

• <strong>The</strong> tables of recommendations for vaccinations in solid organ transplant<br />

(Table 3.3.2) and haematopoietic stem cell transplant (Table 3.3.3) recipients<br />

have been updated to include new vaccines.<br />

• Information on recommendations for persons infected with human<br />

immunodeficiency virus (HIV) now discusses both children and adults and<br />

includes rotavirus, HPV, varicella and zoster vaccines.<br />

• <strong>The</strong> section on vaccination of persons with functional and anatomical<br />

asplenia has been updated to include new vaccines, and now includes a table<br />

(Table 3.3.5) summarising vaccine recommendations in this group.<br />

• <strong>The</strong> section on vaccination of persons with autoimmune diseases has been<br />

expanded to include those undergoing treatment with immunosuppressive<br />

agents and those with Guillain-Barré syndrome and other chronic conditions<br />

(hypopituitarism and metabolic diseases).<br />

• <strong>The</strong> section on vaccination of recent recipients of normal human<br />

immunoglobulin and other blood products has been expanded.<br />

10 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


• <strong>The</strong> section on vaccination of persons with bleeding disorders has been<br />

updated to include new recommendations for when IM injections should be<br />

deferred and advice regarding vaccination of persons with haemophilia.<br />

• <strong>The</strong> table of recommended vaccinations for persons at risk of occupationally<br />

acquired vaccine-preventable diseases (Table 3.3.7) has been updated to<br />

include new occupational groups and recommendations.<br />

• <strong>The</strong> section on vaccination of migrants to Australia has been expanded.<br />

• A new section has been added to provide recommendations for vaccination<br />

for sex industry workers.<br />

Part 4 Vaccine-preventable diseases<br />

4.1 Cholera<br />

• If the interval between primary immunisation and booster dose is more<br />

than 6 months in children aged 2–6 years, or more than 2 years in adults and<br />

children aged >6 years, primary immunisation must be repeated.<br />

4.2 Diphtheria, 4.19 Tetanus and 4.12 Pertussis<br />

• <strong>The</strong> 1st dose of DTPa-containing vaccines due at 2 months of age can be<br />

given as early as 6 weeks of age.<br />

• Advice is provided that an additional dose of pertussis-containing vaccine<br />

can be given in the 2nd year of life (e.g. at 18 months of age) if parents wish<br />

to minimise the likelihood of their child developing pertussis.<br />

• <strong>The</strong> booster dose of DTPa-containing vaccine recommended at 4 years of age<br />

can be given as early as 3.5 years.<br />

• DTPa-containing vaccines can be used for primary or booster doses in<br />

children aged


• For persons undertaking high-risk travel, a 5-yearly booster dose with dT or<br />

dTpa should be considered for protection against tetanus. In other travellers,<br />

a booster dose of tetanus-containing vaccine should be provided if 10 years<br />

have elapsed since the previous dose.<br />

• More information on the definition of ‘tetanus-prone wounds’ is provided,<br />

and the table (Table 4.19.1) on wound management has been updated to<br />

include recommendations for use of tetanus immunoglobulin (TIG) in<br />

immunocompromised persons.<br />

• Information on diphtheria antitoxin is now contained in Part 5 of<br />

the <strong>Handbook</strong>.<br />

4.3 Haemophilus influenzae type b<br />

• Combination Hib-meningococcal C vaccine (Hib-MenCCV) included.<br />

• Hib vaccination recommendations apply to all children, including Aboriginal<br />

and Torres Strait Islander children, as only PRP-T Hib vaccines have been in<br />

use in recent years.<br />

4.4 Hepatitis A<br />

• <strong>The</strong> section on serological testing for hepatitis A prior to vaccination has<br />

been expanded, and more detail provided as to rationale for vaccination of<br />

certain groups.<br />

• Hepatitis A vaccination is recommended in preference to NHIG for use in<br />

post-exposure prophylaxis in immunocompetent persons ≥12 months of age.<br />

4.5 Hepatitis B<br />

• Different schedules for hepatitis B vaccination, including minimum intervals<br />

between doses, have been described in more detail.<br />

• Advice is provided regarding the validity of a hepatitis B vaccine schedule<br />

used for children born overseas, who were vaccinated at birth, 1 month and<br />

6 months of age.<br />

• Information is provided on checking for infection/immunity to hepatitis B<br />

in infants born to mothers with chronic hepatitis B infection 3 to 12 months<br />

after the primary vaccine course.<br />

• It is now recommended that Aboriginal and Torres Strait Islander people<br />

have their risks and vaccination status for hepatitis B reviewed, be offered<br />

testing for previous hepatitis B infection, and be offered vaccination if<br />

non-immune.<br />

• Migrants from hepatitis B endemic countries should be offered testing for<br />

hepatitis B, and vaccination if appropriate.<br />

• <strong>The</strong> section on serological testing for hepatitis B prior to vaccination has<br />

been expanded, and more detail provided as to rationale for testing and/or<br />

vaccination of certain groups, including hepatitis B vaccine non-responders.<br />

12 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.6 Human papillomavirus<br />

• HPV vaccination is now recommended for girls at the optimal age for<br />

vaccination of 11–13 years.<br />

• HPV vaccination is now not routinely recommended for women aged<br />

19–26 years. A risk-benefit assessment should be conducted when<br />

contemplating vaccination of women in this and older age groups.<br />

• Recommendations for use of HPV in males have been included. HPV<br />

vaccination is recommended for males aged 9–18 years, with the optimal<br />

age for vaccination being 11–13 years.<br />

• Specific recommendations regarding the use of HPV vaccine in<br />

immunocompromised persons and men who have sex with men are<br />

now included.<br />

4.7 Influenza<br />

• Intradermal influenza vaccines are included.<br />

• Ages for which different brands of influenza vaccine are registered have been<br />

specified.<br />

• Readers are referred to the Immunise Australia website (www.immunise.<br />

health.gov.au) to check annual statements on influenza vaccine availability<br />

and recommendations for use.<br />

• Information on the disease burden and benefits of influenza vaccination in<br />

pregnancy and in children aged ≥6 months and


• MMR vaccine is to be used for the 1st dose at 12 months of age. MMRV is not<br />

recommended for use as the 1st dose of MMR-containing vaccine in children<br />


• For Aboriginal and Torres Strait Islander children living in the Northern<br />

Territory, Queensland, South Australia or Western Australia, a booster dose of<br />

13vPCV at 12–18 months of age replaces the booster dose of 23vPPV at 18–24<br />

months of age.<br />

• <strong>The</strong> list of conditions associated with increased risks of invasive<br />

pneumococcal disease (IPD) (List 4.13.1) has been revised to include both<br />

adults and children, and is now divided into two categories: those conditions<br />

posing the highest increased risk of developing IPD and those associated with<br />

an increased risk of IPD.<br />

• <strong>The</strong> table (Table 4.13.3) summarising recommendations for vaccination of<br />

adults with 23vPPV has been revised.<br />

• Recommendations for the use of a single dose of 13vPCV in adults and<br />

children >5 years of age with conditions associated with the highest increased<br />

risk of IPD (and who have not previously received a 13vPCV dose) are<br />

included.<br />

• Information on the use of 23vPPV in persons >5 years of age at increased risk<br />

of IPD has also been more clearly presented.<br />

4.14 Poliomyelitis<br />

• <strong>The</strong> 1st dose of IPV-containing combination vaccine due at 2 months of age<br />

can be given as early as 6 weeks of age.<br />

• A booster dose of IPV-containing vaccine is recommended at 4 years of age,<br />

but can be given as early as 3.5 years.<br />

4.15 Q fever<br />

• Q fever vaccination and skin testing training is now undertaken via an<br />

educational module available online.<br />

• Information on the <strong>Australian</strong> Q Fever Register, which lists Q fever<br />

immunisation service providers and records of Q fever vaccinations given to<br />

some persons, is included.<br />

• Q fever vaccination is now also recommended for professional dog and<br />

cat breeders, and wildlife and zoo workers who have contact with at-risk<br />

animals, including kangaroos and bandicoots.<br />

4.16 Rabies and other lyssaviruses (including <strong>Australian</strong> bat lyssavirus)<br />

• <strong>The</strong> terms PEP (post-exposure prophylaxis) and PreP (pre-exposure<br />

prophylaxis) are used throughout the chapter.<br />

• Information and recommendations on management of all potential lyssavirus<br />

exposures, including lyssavirus infection from exposure to bats in non-rabiesenzootic<br />

countries, is now included.<br />

• A 4-dose PEP schedule is now recommended for immunocompetent<br />

persons. A 5-dose schedule is only recommended for persons who are<br />

immunocompromised.<br />

PART 1 INTRODUCTION TO THE AUSTRALIAN IMMUNISATION HANDBOOK 15<br />

1.4 WHAT'S NEW


• A table (Table 4.16.1) summarising World Health Organization (WHO)<br />

categories of lyssavirus exposure, for guidance in use of post-exposure<br />

prophylaxis, has been added.<br />

• Algorithms are provided with details of the recommended management<br />

pathways for post-exposure prophylaxis for rabies and other lyssaviruses<br />

(including <strong>Australian</strong> bat lyssavirus), and for booster doses for persons at<br />

ongoing risk of exposure to rabies and other lyssaviruses.<br />

• Advice regarding the completion of post-exposure prophylaxis commenced<br />

overseas has been expanded, including the addition of a summary table<br />

(Table 4.16.2).<br />

• Persons who have completed a primary course of a currently available cell<br />

culture-derived rabies vaccine no longer routinely require booster doses if<br />

travelling or living in an area of high risk.<br />

• Information on the role of serological testing has been more clearly presented.<br />

4.17 Rotavirus<br />

• <strong>The</strong> upper age limits for each dose of rotavirus vaccines are more clearly<br />

defined.<br />

• Contraindications to rotavirus vaccination now include previous history of<br />

intussusception (IS) and severe combined immunodeficiency in infants.<br />

• Information on the safety of rotavirus vaccines in infants with underlying<br />

conditions and infants who are immunocompromised has been updated.<br />

• Information on adverse events following rotavirus vaccination has been<br />

updated and expanded, including new information on the low, but increased,<br />

risk of IS occurring following the 1st or 2nd dose of either rotavirus vaccine.<br />

4.20 Tuberculosis<br />

• Bacille Calmette-Guérin (BCG) vaccination is no longer routinely<br />

recommended for neonates weighing


Part 5 Passive immunisation<br />

• Information regarding the use of intravenous immunoglobulins as treatment<br />

for disease conditions (such as Kawasaki disease) or as replacement therapy<br />

for immunodeficient individuals is no longer included in the <strong>Handbook</strong>.<br />

Readers are referred to National Blood Authority guidelines.<br />

PART 1 INTRODUCTION TO THE AUSTRALIAN IMMUNISATION HANDBOOK 17<br />

1.4 WHAT'S NEW


1.5 FUNDAMENTALS OF IMMUNISATION<br />

1.5.1 Overview<br />

Vaccines are complex biological products designed to induce a protective<br />

immune response effectively and safely. Vaccines contain one or more antigens<br />

(or immunogens) that stimulate an active immune response. <strong>The</strong>se are generally<br />

protein- or polysaccharide (sugar)-based substances. <strong>The</strong> number and derivation<br />

of the antigen(s) contained in each vaccine vary. Most vaccines work by inducing<br />

B-lymphocytes to produce antibodies that bind to and inhibit pathogenic<br />

organisms or their toxins. Generation of T-cell-mediated (cellular) immunity is<br />

also important for some vaccines.<br />

Vaccines, like all medicines, are regulated in Australia by the <strong>The</strong>rapeutic Goods<br />

Administration (TGA). Before they are made available for use they are rigorously<br />

tested in human clinical trials to confirm that they are safe and that they<br />

stimulate protective immune responses. Vaccines are also evaluated to ensure<br />

compliance with strict manufacturing and production standards. This testing is<br />

required by law and is usually conducted both during the vaccine’s development<br />

and after its registration. In addition, once they are in use, the safety of vaccines<br />

is monitored by the TGA and other organisations using different methods,<br />

including passive and active surveillance for adverse events following<br />

immunisation (see 1.5.5 Vaccine safety and adverse events following immunisation).<br />

1.5.2 Passive immunisation<br />

Passive immunity is the direct transfer or administration of antibodies to a<br />

non-immune person to provide immediate protection. One example of passive<br />

immunisation is the transfer of maternal antibodies to the fetus, which provides<br />

some short-lived protection of the newborn infant against certain infections. 1,2<br />

Another example is the administration of a product containing antibodies<br />

(or immunoglobulins, IgG) pooled from blood donors, in order to provide<br />

temporary protection to a non-immune person who has recently been exposed to<br />

infection. 3 <strong>The</strong> protection afforded is immediate, but lasts for only a few weeks<br />

as the half-life of IgG is approximately 3 to 4 weeks. Regular immunoglobulin<br />

infusions are also indicated for some immunocompromised persons who are<br />

deficient in antibody. A separate use of immunoglobulins is in the treatment<br />

of a number of specific immune-mediated conditions in order to modulate<br />

the disease course. For further information regarding the use of intravenous<br />

immunoglobulin for this purpose, refer to Criteria for the clinical use of intravenous<br />

immunoglobulin in Australia (www.nba.gov.au/ivig/index.html).<br />

For more information on passive immunisation see Part 5 Passive immunisation.<br />

1.5.3 Active immunisation<br />

Active immunisation involves the use of vaccines to stimulate the immune<br />

system to produce a protective immune response. Vaccines usually induce<br />

an immune response that mimics the host’s response to natural infection, but<br />

18 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


without the harmful consequences of the infection itself. In addition to antibody<br />

responses, many vaccines also stimulate cell-mediated immunity. Immunity<br />

following active immunisation generally lasts for months to many years,<br />

depending on the nature of the vaccine as well as host factors. 4,5 Protective<br />

immunity is induced by antigen(s) contained within the vaccine. This may be a<br />

toxoid (a bacterial toxin that has been rendered non-toxigenic, e.g. for tetanus or<br />

diphtheria); killed or inactivated bacteria or viruses, such as hepatitis A vaccines;<br />

live attenuated bacteria or viruses, such as measles, mumps and rubella vaccines;<br />

or subunit components of a pathogen that only contain the antigen(s) of interest,<br />

such as the hepatitis B vaccine. 4,5<br />

In addition to containing the immunising antigen(s), vaccines may also contain<br />

the following:<br />

• Adjuvants, which enhance the immune response to an antigen; an example is<br />

aluminium hydroxide.<br />

• Preservatives, which reduce the risk of contamination; some examples<br />

are 2-phenoxyethanol, which is also used in many cosmetics and<br />

pharmaceuticals, and thiomersal, which is used in the Q fever vaccine but is<br />

not present in any of the vaccines on the National <strong>Immunisation</strong> Program for<br />

young children.<br />

• Stabilisers, which improve the shelf-life and help to protect the vaccine from<br />

adverse conditions; examples are sucrose, mannitol, lactose and gelatin.<br />

Stabilisers are also used in most confectionery and many pharmaceuticals.<br />

• Emulsifiers or surfactants, which alter the surface tension of the liquid<br />

vaccine; examples are polysorbate-80 and sorbitol. Emulsifiers are added to<br />

most ice creams and many pharmaceuticals.<br />

• Residuals, which are minute or trace amounts of substances that remain after<br />

the manufacture of the vaccine; examples of residuals detectable in some<br />

vaccines are formaldehyde, antibiotics such as neomycin or polymyxin, and<br />

egg proteins.<br />

Further details of a particular vaccine’s constituents can be found in either the<br />

product information (PI) or the consumer medicines information (CMI) for<br />

individual vaccines. This information is presented in the shaded box for each<br />

vaccine under the disease-specific chapters in Part 4 of this <strong>Handbook</strong> (current<br />

June 2012); however, it is important to note that PIs and CMIs are updated<br />

periodically. <strong>The</strong> most current versions of the PI (and CMI) for vaccines (and<br />

other medicines) are available from the TGA website (www.tga.gov.au).<br />

In addition, information on the components contained in vaccines that are<br />

available under the <strong>Australian</strong> National <strong>Immunisation</strong> Program is provided in<br />

Appendix 3 of this <strong>Handbook</strong>, and further details on vaccine composition can be<br />

found in Appendix 4 Commonly asked questions about vaccination.<br />

PART 1 INTRODUCTION TO THE AUSTRALIAN IMMUNISATION HANDBOOK 19<br />

1.5 FUNDAMENTALS<br />

OF IMMUNISATION


<strong>The</strong> recommended number of doses and age of administration vary for each<br />

vaccine. <strong>The</strong>se recommendations are based on the type of vaccine, disease<br />

epidemiology (the age-specific risk for infection and for complications), and the<br />

anticipated immune response of the recipient (including whether transplacental<br />

transfer of maternal antibodies will inhibit the immune response in an infant). 4,5<br />

Several doses of a vaccine may be required to induce protective immunity,<br />

particularly in younger children.<br />

Homeopathic preparations do not induce immunity and are never an alternative<br />

to vaccination (see Appendix 4 Commonly asked questions about vaccination).<br />

Detailed information on the background, available vaccines and<br />

recommendations for vaccines used in active immunisation are provided in the<br />

disease-specific chapters in Part 4 of this <strong>Handbook</strong>.<br />

1.5.4 Vaccine efficacy, vaccine effectiveness and vaccine failure<br />

<strong>The</strong> terms vaccine efficacy and vaccine effectiveness are often used<br />

interchangeably. However, in general terms, vaccine efficacy refers to estimates of<br />

protection obtained under the idealised conditions of a randomised controlled<br />

trial (RCT). It is usually expressed as the percentage reduction in a person’s<br />

risk of disease if vaccinated compared to the risk if not vaccinated. Vaccine<br />

effectiveness refers to estimates of protection obtained under ‘real world’ rather<br />

than trial conditions, for example, in immunisation programs after vaccine<br />

registration. Sometimes vaccine effectiveness is also taken to include the<br />

broader impact of a vaccination program on overall disease incidence in the<br />

population, including any additional herd protection conferred to unvaccinated<br />

individuals. 4,6<br />

<strong>The</strong> extent and duration of protection provided by vaccination varies and<br />

is influenced by many factors. For example, some vaccines, such as the<br />

pneumococcal and meningococcal polysaccharide vaccines, provide protection<br />

for a few years only. This is because polysaccharide antigens induce antibodies<br />

without the involvement of T-lymphocytes (T-cell independent response). T-cell<br />

lymphocyte involvement is needed for long-term immune memory; without it,<br />

protection is relatively short-lived and immunity wanes, sometimes requiring<br />

revaccination. In addition, polysaccharide vaccines are less immunogenic<br />

in children aged


(vaccine failure). Often such infections result in a milder or more attenuated form<br />

of disease, for example, chickenpox developing despite varicella vaccination or<br />

whooping cough developing after 2 or more doses of pertussis vaccine. Vaccine<br />

failure can be categorised in two ways. ‘Primary’ vaccine failure occurs when<br />

a fully vaccinated person does not form an adequate immune response to that<br />

vaccine. This might occur because a vaccine is defective due to a manufacturing<br />

fault or, more typically, because of inadequate storage (e.g. breakage of the cold<br />

chain) or expiry of the shelf-life. Primary vaccine failure may also occur because<br />

the recipient’s immune response is ineffective, which may be relatively specific<br />

for that vaccine or part of a broader immunodeficiency. ‘Secondary’ vaccine<br />

failures occur when a fully vaccinated person becomes susceptible to that disease<br />

over time, usually because immunity following vaccination wanes over time.<br />

As discussed above, the duration of the protective effect of vaccination varies<br />

depending on the nature of the vaccine and the type of immune response elicited,<br />

the number of doses received, and host factors. Some vaccinated persons may get<br />

further immune stimulation from natural infection or colonisation, which aids in<br />

maintaining ongoing protection.<br />

1.5.5 Vaccine safety and adverse events following immunisation<br />

What is an adverse event following immunisation?<br />

<strong>The</strong> term ‘adverse event following immunisation’ (AEFI) refers to any<br />

untoward medical occurrence that follows immunisation, whether<br />

expected or unexpected, and whether triggered by the vaccine or only<br />

coincidentally occurring after receipt of a vaccine. <strong>The</strong> adverse event may<br />

be any unfavourable or unintended sign, abnormal laboratory finding,<br />

symptom or disease. 7<br />

Adverse events following immunisation (AEFI) should be reported<br />

promptly, either according to relevant state or territory protocols or directly<br />

to the TGA (for detailed information on reporting and management of AEFI,<br />

see 2.3 Post-vaccination).<br />

<strong>The</strong> safety of vaccines is very important as vaccines are given to prevent disease<br />

and target all or many members of the population, most of whom are healthy.<br />

All vaccines available in Australia must pass stringent safety testing before being<br />

approved for use by the TGA. This testing is required by law and is usually<br />

done over many years during the vaccine’s development. In addition, the TGA<br />

monitors the safety of vaccines once they are registered.<br />

From the time a vaccine comes into use, there is ongoing review of both vaccine<br />

safety and efficacy through a variety of mechanisms, such as further clinical<br />

PART 1 INTRODUCTION TO THE AUSTRALIAN IMMUNISATION HANDBOOK 21<br />

1.5 FUNDAMENTALS<br />

OF IMMUNISATION


trials and surveillance of disease and vaccine adverse events. One important<br />

component of ensuring that vaccines are safe is to monitor the occurrence of<br />

AEFI. In Australia, there are regional and national surveillance systems that<br />

collect reports of any adverse events following immunisation. All AEFI reported<br />

are added to the national Adverse Drug Reactions System (ADRS) database,<br />

which is operated by the TGA. (See also 2.3 Post-vaccination.) Each year, reports<br />

presenting data and analysis of AEFI in Australia are published in the journal<br />

Communicable Diseases Intelligence, accessible via the <strong>Australian</strong> Government<br />

Department of Health and Ageing website (www.health.gov.au/internet/main/<br />

publishing.nsf/content/cda-pubs-cdi-cdiintro.htm).<br />

In some cases, other specific studies will be conducted to ensure that vaccine<br />

safety is closely monitored once a vaccine is in use. For example, the risk of<br />

intussusception (IS) following rotavirus vaccines has been closely monitored<br />

in Australia and elsewhere because of the association of a previously licensed<br />

vaccine with an unacceptably high risk of IS.<br />

Adverse reactions to vaccines (also known as ‘vaccine side effects’) do sometimes<br />

occur. It is usually not possible to predict which individuals may have a mild<br />

or, rarely, a serious reaction to a vaccine. However, by following guidelines<br />

regarding when vaccines should and should not be used, the risk of adverse<br />

events can be minimised. As vaccines are usually given to healthy people, any<br />

adverse event that follows soon after immunisation may be perceived as due to<br />

the vaccine. <strong>The</strong> fact that an adverse event occurs after an immunisation does not<br />

prove the vaccine caused the event. A causal association is rarely certain, but is<br />

most likely when the AEFI is both typical (even if very rare) and when there is<br />

no other plausible explanation, for example, an injection site reaction occurring<br />

a day after vaccination or typical anaphylaxis occurring within minutes of<br />

vaccination. Many AEFIs are less specific and/or have plausible alternative<br />

explanations, including coincidence. Such associations can only be assessed by<br />

large-scale epidemiological studies or specific tests, for example, in the case of<br />

allergy, by allergy testing or challenge. Even when an AEFI is typical, it may be<br />

nonetheless unrelated to vaccination (see 2.3 Post-vaccination).<br />

Vaccine adverse events fall into two general categories: local or systemic. Local<br />

reactions are defined as reactions occurring at the site of vaccine administration<br />

(usually pain, redness or swelling at the injection site) and are generally the least<br />

severe and most frequently occurring AEFI. Systemic reactions most commonly<br />

include fever, headache and lethargy. 8,9 Allergic reactions can also occur, although<br />

anaphylaxis, the most severe form of an allergic response, is rarely caused by<br />

vaccination. It is not possible to completely predict which individuals may have a<br />

reaction to a vaccine.<br />

Each chapter in the <strong>Handbook</strong> indicates under which circumstances vaccine<br />

administration is contraindicated or where precautions are required. A<br />

contraindication to vaccination usually occurs when a person has a pre-existing<br />

condition that significantly increases the chance that a serious adverse event<br />

22 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


will occur following receipt of a specific vaccine. A contraindication may also<br />

occur when there is insufficient safety data regarding a vaccine’s use and there<br />

is a theoretical risk of harm. In general, vaccines should not be given where a<br />

contraindication exists, except under advice from your local state or territory<br />

health department (Appendix 1 Contact details for <strong>Australian</strong>, state and territory<br />

government health authorities and communicable disease control).<br />

A precaution is a condition that may increase the chance of an adverse event<br />

following immunisation or one that may compromise the ability of the vaccine<br />

to produce immunity. When a precaution exists, there may still be circumstances<br />

when the benefits of giving the vaccine outweigh the potential risks; however,<br />

special care and the provision of appropriate advice to the vaccine recipient may<br />

be required (see 3.3.1 Vaccination of persons who have had an adverse event following<br />

immunisation).<br />

In 2010, a national review of the management of adverse events that occurred<br />

following influenza vaccine administration was performed. 10 <strong>The</strong> review made a<br />

number of recommendations to further improve the monitoring of vaccine safety<br />

in Australia. Any changes to the system(s) for monitoring or reporting of AEFI<br />

in Australia will be reflected in future updates to the <strong>Handbook</strong> and will also be<br />

available from the Immunise Australia website (www.immunise.health.gov.au).<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

PART 1 INTRODUCTION TO THE AUSTRALIAN IMMUNISATION HANDBOOK 23<br />

1.5 FUNDAMENTALS<br />

OF IMMUNISATION


PART 2 VACCINATION PROCEDURES<br />

In Australia, vaccination is undertaken predominantly through general practices,<br />

but in some jurisdictions vaccines may be given through local council clinics,<br />

community centres or through school-based immunisation programs. In some<br />

situations, vaccinations may also be given in travel medicine clinics, public<br />

hospitals, staff occupational health clinics and aged care facilities. State or<br />

territory legislation outlines who can access and administer vaccines. All vaccines<br />

must be administered in accordance with the relevant legislation, best practice<br />

and the following <strong>Handbook</strong> guidelines and recommendations.<br />

2.1 PRE-VACCINATION<br />

<strong>The</strong> following sections discuss steps and procedures that should occur before a<br />

vaccination encounter.<br />

2.1.1 Preparing an anaphylaxis response kit<br />

<strong>The</strong> availability of protocols, equipment and drugs necessary for the<br />

management of anaphylaxis should be checked before each vaccination session.<br />

An anaphylaxis response kit should be on hand at all times and should contain:<br />

• adrenaline 1:1000 (minimum of three ampoules – check expiry dates)<br />

• minimum of three 1 mL syringes and 25 mm length needles (for<br />

intramuscular [IM] injection)<br />

• cotton wool swabs<br />

• pen and paper to record time of administration of adrenaline<br />

• laminated copy of adrenaline doses (Table 2.3.2 or back cover of this<br />

<strong>Handbook</strong>)<br />

• laminated copy of ‘Recognition and treatment of anaphylaxis’ (back cover<br />

of this <strong>Handbook</strong>).<br />

See 2.3.2 Adverse events following immunisation for details on recognition and<br />

treatment of adverse events following immunisation (in particular, see ‘Use of<br />

adrenaline’ and ‘Use of adrenaline autoinjectors for anaphylaxis treatment’ in<br />

that section).<br />

2.1.2 Effective cold chain: transport, storage and handling of vaccines<br />

<strong>The</strong> cold chain is the system of transporting and storing vaccines within the<br />

temperature range of +2°C to +8°C from the place of manufacture to the point<br />

of administration. 1 Maintenance of the cold chain is essential for maintaining<br />

vaccine potency and, in turn, vaccine effectiveness. This is vital, not only for<br />

those vaccines provided as part of the National <strong>Immunisation</strong> Program, but also<br />

for vaccines purchased by the patient via prescription from a pharmacist. In such<br />

24 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


cases, both the doctor issuing the prescription and the pharmacist dispensing<br />

the vaccine must inform the patient of the need for maintaining, and how to<br />

maintain, the cold chain for the vaccine they have purchased.<br />

All immunisation service providers must be familiar with, and adhere<br />

to, the National vaccine storage guidelines: Strive for 5. 1 This publication can<br />

be accessed free of charge from www.immunise.health.gov.au/internet/<br />

immunise/publishing.nsf/Content/provider-store<br />

<strong>The</strong> National vaccine storage guidelines: Strive for 5 contains specific details on<br />

setting up the infrastructure for a vaccination service, and immunisation service<br />

providers should refer to this document to ensure that satisfactory equipment<br />

and procedures are in place before commencing vaccination services. 1<br />

<strong>The</strong>se guidelines also provide instructions on how best to transport vaccines<br />

from the main storage facility to outreach or external clinics. Purpose-built<br />

vaccine refrigerators (PBVR) are the preferred means of storage for vaccines.<br />

Domestic refrigerators are not designed for the special temperature needs of<br />

vaccine storage.<br />

Cold chain breaches<br />

Despite best practices, cold chain breaches sometimes occur. It is important<br />

to report any cold chain breaches so that revaccination of patients or recall of<br />

unused vaccines can be undertaken, if required.<br />

Do not discard or use any vaccines exposed to temperatures below +2°C or<br />

above +8°C without obtaining further advice. Isolate vaccines and contact<br />

the state/territory health authorities (see Appendix 1) for advice on the<br />

National <strong>Immunisation</strong> Program vaccines and the manufacturer/supplier<br />

for privately purchased vaccines. Recommendations for the discarding of<br />

vaccines may differ between health authorities and manufacturers.<br />

2.1.3 Valid consent<br />

Valid consent can be defined as the voluntary agreement by an individual to a<br />

proposed procedure, given after sufficient, appropriate and reliable information<br />

about the procedure, including the potential risks and benefits, has been<br />

conveyed to that individual. 2-6 As part of the consent procedure, persons to be<br />

vaccinated and/or their parents/carers should be given sufficient information<br />

PART 2 VACCINATION PROCEDURES 25<br />

2.1 PRE-VACCINATION


(preferably written) on the risks and benefits of each vaccine, including what<br />

adverse events are possible, how common they are and what they should do<br />

about them7 (the table inside the front cover of this <strong>Handbook</strong>, Side effects following<br />

immunisation for vaccines used in the National <strong>Immunisation</strong> Program (NIP) schedule,<br />

can be used for this purpose).<br />

For consent to be legally valid, the following elements must be present: 6,8<br />

1. It must be given by a person with legal capacity, and of sufficient intellectual<br />

capacity to understand the implications of being vaccinated.<br />

2. It must be given voluntarily in the absence of undue pressure, coercion or<br />

manipulation.<br />

3. It must cover the specific procedure that is to be performed.<br />

4. It can only be given after the potential risks and benefits of the relevant<br />

vaccine, risks of not having it and any alternative options have been<br />

explained to the individual.<br />

<strong>The</strong> individual must have sufficient opportunity to seek further details or<br />

explanations about the vaccine(s) and/or its administration. <strong>The</strong> information<br />

must be provided in a language or by other means the individual can<br />

understand. Where appropriate, an interpreter and/or cultural support person<br />

should be involved.<br />

Consent should be obtained before each vaccination, once it has been established<br />

that there are no medical condition(s) that contraindicate vaccination. Consent<br />

can be verbal or written. <strong>Immunisation</strong> providers should refer to their state or<br />

territory’s policies on obtaining written consent (see Appendix 1 Contact details<br />

for <strong>Australian</strong>, state and territory government health authorities and communicable<br />

disease control).<br />

Consent on behalf of a child or adolescent<br />

In general, a parent or legal guardian of a child has the authority to consent to<br />

vaccination of that child; however, it is important to check with your state or<br />

territory authority where any doubt exists. 2,5 A child in this context is defined<br />

as being under the age of 18 years in Tasmania, Victoria and Western Australia;<br />

under the age of 14 years in New South Wales; and under the age of 16 years<br />

in the <strong>Australian</strong> Capital Territory, South Australia and the Northern Territory.<br />

Queensland follows common law principles.<br />

For certain procedures, including vaccination, persons younger than the ages<br />

defined above may have sufficient maturity to understand the proposed<br />

procedure and the risks and benefits associated with it, and thus may have the<br />

capacity to consent under certain circumstances. Refer to the relevant state or<br />

territory immunisation service provider guidelines for more information.<br />

26 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Should a child or adolescent refuse a vaccination for which a parent/guardian<br />

has given consent, the child/adolescent’s wishes should be respected and the<br />

parent/guardian informed. 2<br />

Consent on behalf of an adult lacking capacity<br />

A careful assessment should be made of an adult’s capacity to give valid consent<br />

to a vaccination. If the adult lacks capacity, practitioners should refer to relevant<br />

state and territory laws relating to obtaining consent from a substitute decisionmaker.<br />

For example, this may occur for influenza vaccination of an elderly person<br />

with dementia. Refer to the enduring guardianship legislation appropriate for<br />

your state or territory for further advice.<br />

Resources to help communicate the risks and benefits of vaccines<br />

Plain language should be used when communicating information about vaccines<br />

and their use. <strong>The</strong> person to be vaccinated (or their parent/guardian) must be<br />

encouraged and allowed to ask for further information and have sufficient time<br />

to make a decision about whether to consent or not. 9,10<br />

It is preferable that printed information is available to supplement any verbal<br />

explanations. 11 <strong>The</strong> summary table Comparison of the effects of diseases and the side<br />

effects of NIP vaccines inside the back cover of this <strong>Handbook</strong> provides some basic<br />

information necessary to communicate the risks and benefits of vaccination.<br />

<strong>The</strong> table can be photocopied and used freely as required.<br />

More detailed information concerning vaccines and their use is available from<br />

the following sources:<br />

• www.immunise.health.gov.au<br />

<strong>The</strong> Immunise Australia website includes the booklet Understanding childhood<br />

immunisation, which contains frequently asked questions and links to<br />

state and territory health department websites. Several of these sites offer<br />

multilingual fact sheets.<br />

• www.ncirs.edu.au<br />

<strong>The</strong> National Centre for <strong>Immunisation</strong> Research and Surveillance of Vaccine<br />

Preventable Diseases website includes fact sheets related to specific vaccines,<br />

vaccine-preventable diseases and vaccine safety. <strong>The</strong> website also hosts<br />

online decision aids to assist patients in deciding whether to vaccinate or not.<br />

See also Appendix 4 Commonly asked questions about vaccination.<br />

Evidence of consent<br />

General practice or public immunisation clinics<br />

Consent may be given either in writing or verbally, according to the protocols<br />

of the health facility, but it must meet the criteria for valid consent. Evidence<br />

of verbal consent should be documented in the clinical records. If a standard<br />

procedure is routinely followed in a practice or clinic, then a stamp, a sticker<br />

PART 2 VACCINATION PROCEDURES 27<br />

2.1 PRE-VACCINATION


or a provider’s signature indicating that the routine procedure has been<br />

followed may be used. For paperless medical records, a typed record of<br />

verbal consent may be made in the patient’s file, or a copy of written consent<br />

scanned into the file.<br />

Explicit verbal consent is required before administration of any vaccine, even<br />

when written consent has been given at previous vaccination encounters for the<br />

same vaccine. Verbal consent should be documented in the patient’s file each<br />

time it is given.<br />

School-based vaccination programs<br />

Consent is required for provision of individual vaccines or a vaccine course<br />

offered in school-based vaccination programs.<br />

In school-based, and other large-scale, vaccination programs, the parent or<br />

guardian usually does not attend with the child on the day the vaccination is<br />

given, and written consent from the parent or guardian is desirable in these<br />

circumstances. However, if written consent is not able to be provided, or if<br />

further clarification is required, verbal consent may be sought by telephone<br />

from the parent or guardian by the immunisation service provider. This should<br />

be clearly documented on the child’s consent form. In some jurisdictions,<br />

older adolescents may be able to provide their own consent for vaccinations<br />

offered through school-based vaccination programs. 12 Consent requirements<br />

and vaccines offered in these programs vary between jurisdictions. Refer to the<br />

relevant state or territory school-based vaccination program guidelines for more<br />

information.<br />

2.1.4 Pre-vaccination screening<br />

<strong>Immunisation</strong> service providers should perform a comprehensive prevaccination<br />

health screen of all persons to be vaccinated. For some individuals,<br />

alterations to the routinely recommended vaccines may be necessary to either<br />

eliminate or minimise the risk of adverse events, to optimise an individual’s<br />

immune response, or to enhance the protection of a household contact against<br />

vaccine-preventable diseases.<br />

Providers should:<br />

• ensure that they have the right person to be vaccinated<br />

• ensure which vaccine(s) are indicated, including any previously<br />

missed vaccine doses<br />

• consider whether alternative or additional vaccines should be given<br />

• check if there are any contraindications or precautions to the vaccines<br />

that are to be given<br />

• ensure that the patient to be vaccinated is the appropriate age for the<br />

vaccines to be given<br />

28 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


• check that the correct time interval has passed since any previous vaccine(s)<br />

or any blood products were given.<br />

See also 2.1.5 Catch-up and relevant disease chapters for further details.<br />

Steps for pre-vaccination screening<br />

Follow these steps to complete the pre-vaccination screening process:<br />

• Provide the person to be vaccinated or the parent/carer with the Prevaccination<br />

screening checklist (Table 2.1.1).<br />

» Some of the questions in this checklist are deliberately non-specific so as to<br />

elicit as much important information as possible.<br />

» <strong>The</strong> checklist may be photocopied and handed to the person to be<br />

vaccinated or the parent/carer just before vaccination.<br />

» <strong>The</strong> checklist may also be photocopied and displayed in the clinic/surgery<br />

for easy reference for the immunisation service provider.<br />

• For vaccination of adults, seek additional information about the occupation<br />

and lifestyle factors that may influence vaccination requirements. This is<br />

discussed in more detail in 2.1.5 Catch-up below under ‘Catch-up schedules<br />

for persons ≥10 years of age’.<br />

• If you identify the presence of a condition or circumstance indicated on the<br />

pre-vaccination screening checklist, refer to Table 2.1.2, which lists the specific<br />

issues pertaining to such condition(s) or circumstances and provides the<br />

appropriate action with a rationale.<br />

• Where necessary, seek further advice from a specialist immunisation clinic,<br />

a medical practitioner with expertise in vaccination, the immunisation<br />

section within your state or territory health authority, or your local Public<br />

Health Unit (see Appendix 1 Contact details for <strong>Australian</strong>, state and territory<br />

government health authorities and communicable disease control).<br />

PART 2 VACCINATION PROCEDURES 29<br />

2.1 PRE-VACCINATION


Table 2.1.1: Pre-vaccination screening checklist<br />

Pre-vaccination screening checklist<br />

This checklist helps decide about vaccinating you or your child today. Please<br />

fill in the following information for your doctor/nurse.<br />

Name of person to be vaccinated ______________________________________<br />

Date of birth ________________<br />

Age today __________________<br />

Name of person completing this form __________________________________<br />

Please indicate if the person to be vaccinated:<br />

is unwell today<br />

has a disease that lowers immunity (e.g. leukaemia, cancer, HIV/AIDS) or is<br />

having treatment that lowers immunity (e.g. oral steroid medicines such as<br />

cortisone and prednisone, radiotherapy, chemotherapy)<br />

has had a severe reaction following any vaccine<br />

has any severe allergies (to anything)<br />

has had any vaccine in the past month<br />

has had an injection of immunoglobulin, or received any blood products or<br />

a whole blood transfusion within the past year<br />

is pregnant<br />

has a past history of Guillain-Barré syndrome<br />

was a preterm infant<br />

has a chronic illness<br />

has a bleeding disorder<br />

identifies as an Aboriginal or Torres Strait Islander<br />

does not have a functioning spleen<br />

is planning a pregnancy or anticipating parenthood<br />

is a parent, grandparent or carer of a newborn<br />

lives with someone who has a disease that lowers immunity (e.g. leukaemia,<br />

cancer, HIV/AIDS), or lives with someone who is having treatment<br />

that lowers immunity (e.g. oral steroid medicines such as cortisone and<br />

prednisone, radiotherapy, chemotherapy)<br />

is planning travel<br />

has an occupation or lifestyle factor(s) for which vaccination may be needed<br />

(discuss with doctor/nurse)<br />

Please specify:__________________________________________________<br />

30 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Note: Please discuss this information or any questions you have about<br />

vaccination with your doctor/nurse before the vaccines are given.<br />

Before any vaccination takes place, your doctor/nurse should ask you:<br />

Did you understand the information provided to you about vaccination?<br />

Do you need more information to decide whether to proceed?<br />

Did you bring your/your child’s vaccination record card with you?<br />

It is important for you to receive a personal record of your or your child’s<br />

vaccinations. If you do not have a record, ask your doctor/nurse to give<br />

you one. Bring this record with you every time you or your child visit for<br />

vaccination. Make sure your doctor/nurse records all vaccinations on it.<br />

Conditions or circumstances identified using the pre-vaccination<br />

screening checklist<br />

<strong>The</strong> recommended responses for immunisation service providers to make if any<br />

conditions or circumstances are identified by using the pre-screening checklist<br />

are summarised in Table 2.1.2.<br />

Note: Only vaccines recommended on the NIP schedule are included in Table<br />

2.1.2. For information on other vaccines, refer to the relevant disease-specific<br />

chapter in Part 4 of this <strong>Handbook</strong> or to vaccine product information.<br />

For reference, Table 2.1.3 provides a list of live attenuated vaccines.<br />

Table 2.1.2: Responses to relevant conditions or circumstances identified<br />

through the pre-vaccination screening checklist<br />

Condition or<br />

circumstance of person<br />

to be vaccinated<br />

Is unwell today:<br />

• acute febrile illness<br />

(current T ≥38.5°C)<br />

• acute systemic<br />

illness<br />

Action Rationale 13-15<br />

Defer all vaccines until<br />

afebrile.<br />

Note: Children with minor<br />

illnesses (without acute<br />

systemic symptoms/signs)<br />

should be vaccinated.<br />

To avoid an adverse event<br />

in an already unwell child,<br />

or to avoid attributing<br />

symptoms to vaccination<br />

PART 2 VACCINATION PROCEDURES 31<br />

2.1 PRE-VACCINATION


Condition or<br />

circumstance of person<br />

to be vaccinated<br />

Has a disease that<br />

lowers immunity or is<br />

receiving treatment that<br />

lowers immunity<br />

Has had anaphylaxis<br />

following a previous<br />

dose of the relevant<br />

vaccine<br />

Has a severe allergy to a<br />

vaccine component<br />

Has received a live<br />

attenuated viral<br />

parenteral vaccine* or<br />

BCG vaccine in past 4<br />

weeks<br />

Action Rationale 13-15<br />

Refer to 3.3.3 Vaccination<br />

of immunocompromised<br />

persons.<br />

In some cases, expert<br />

advice may need to be<br />

sought before vaccination<br />

(see Appendix 1).<br />

Note: Persons living with<br />

someone with lowered<br />

immunity should be<br />

vaccinated, including with<br />

live viral vaccines (see<br />

below).<br />

Do not vaccinate. Seek<br />

further medical advice<br />

to confirm causality<br />

and to assist with other<br />

vaccinations.<br />

See also ‘Contraindications<br />

to vaccination’ below.<br />

Refer to Appendix 3 for<br />

a vaccine component<br />

checklist.<br />

Do not vaccinate but seek<br />

specialist advice (see<br />

Appendix 1). <strong>The</strong> patient<br />

may still be able to be<br />

vaccinated, dependent on<br />

the allergy.<br />

Delay live attenuated<br />

viral parenteral vaccines<br />

by 4 weeks.<br />

32 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

<strong>The</strong> safety and<br />

effectiveness of the vaccine<br />

may be suboptimal<br />

in persons who are<br />

immunocompromised.<br />

Live attenuated vaccines<br />

may be contraindicated.<br />

Anaphylaxis to a<br />

previous dose of vaccine<br />

is a contraindication to<br />

receiving the same vaccine.<br />

Anaphylaxis to a vaccine<br />

component is generally<br />

a contraindication to<br />

receiving the vaccine.<br />

<strong>The</strong> immune response<br />

to a live attenuated<br />

viral vaccine (given<br />

parenterally) may interfere<br />

with the response to a<br />

subsequent live viral<br />

vaccine given within 4<br />

weeks of the first.


Condition or<br />

circumstance of person<br />

to be vaccinated<br />

Has had any blood<br />

product in the past<br />

7 months, or has<br />

had IM or IV<br />

immunoglobulin<br />

in the past year<br />

Is planning a pregnancy<br />

or anticipating<br />

parenthood<br />

Action Rationale 13-15<br />

Check which product<br />

the person received<br />

and the interval since<br />

administration. Refer to<br />

Table 3.3.6 Recommended<br />

intervals between either<br />

immunoglobulins or blood<br />

products and MMR, MMRV<br />

or varicella vaccination.<br />

If not eligible, make a<br />

return appointment for this<br />

vaccination, and send a<br />

reminder later if necessary.<br />

Ensure women planning<br />

pregnancy and household<br />

members have received<br />

vaccines recommended<br />

for their age group. For<br />

example, 2nd dose of<br />

MMR † (if born after 1966);<br />

varicella; dTpa; ‡ and/<br />

or have had appropriate<br />

pre-conception serological<br />

testing.<br />

See 3.3.2 Vaccination of<br />

women who are planning<br />

pregnancy, pregnant or<br />

breastfeeding, and preterm<br />

infants.<br />

Advise women not to<br />

become pregnant within<br />

28 days of receiving live<br />

viral vaccines.*<br />

Antibodies in these<br />

products may interfere<br />

with the immune response<br />

to MMR, MMRV and<br />

varicella vaccines.<br />

<strong>The</strong> recommended<br />

interval to vaccination<br />

varies depending on the<br />

immunoglobulin or blood<br />

product administered.<br />

Vaccinating before<br />

pregnancy may prevent<br />

maternal illness, which<br />

could affect the infant,<br />

and may confer passive<br />

immunity to the newborn.<br />

PART 2 VACCINATION PROCEDURES 33<br />

2.1 PRE-VACCINATION


Condition or<br />

circumstance of person<br />

to be vaccinated<br />

Action Rationale 13-15<br />

Is pregnant Refer to Table 3.3.1<br />

Recommendations for<br />

vaccination in pregnancy.<br />

Has a history of<br />

Guillain-Barré syndrome<br />

(GBS)<br />

Influenza vaccine is<br />

recommended for all<br />

pregnant women.<br />

Live vaccines* should<br />

be deferred until after<br />

delivery.<br />

Vaccination of household<br />

contacts of pregnant<br />

women should be<br />

completed according to the<br />

NIP schedule.<br />

See 3.3.3 Vaccination of<br />

immunocompromised persons<br />

and 4.7 Influenza.<br />

Risks and benefits of<br />

influenza vaccine should<br />

be weighed against the<br />

potential risk of GBS<br />

recurrence (seek further<br />

advice as per Appendix 1).<br />

Was born preterm See 3.3.2 Vaccination of<br />

women who are planning<br />

pregnancy, pregnant or<br />

breastfeeding, and preterm<br />

infants.<br />

Preterm infants born at<br />


Condition or<br />

circumstance of person<br />

to be vaccinated<br />

Has a severe or<br />

chronic illness<br />

Action Rationale 13-15<br />

See 3.3 Groups with special<br />

vaccination requirements.<br />

<strong>The</strong>se persons should<br />

receive recommended<br />

vaccines such as<br />

pneumococcal vaccine<br />

and annual influenza<br />

vaccination.<br />

If there is significant<br />

immunocompromise, they<br />

should not receive live<br />

vaccines* (see above).<br />

Has a bleeding disorder See 3.3.5 Vaccination<br />

of persons with bleeding<br />

disorders.<br />

<strong>The</strong> subcutaneous route<br />

could be considered<br />

as an alternative to the<br />

intramuscular route; seek<br />

specialist advice (see<br />

Appendix 1).<br />

Identifies as an<br />

Aboriginal or Torres<br />

Strait Islander<br />

Does not have a<br />

functioning spleen<br />

See 3.1 Vaccination for<br />

Aboriginal and Torres Strait<br />

Islander people.<br />

See the National<br />

<strong>Immunisation</strong> Program for<br />

specific recommendations<br />

for Aboriginal and Torres<br />

Strait Islander people.<br />

See 3.3.3 Vaccination<br />

of immunocompromised<br />

persons, ‘Persons with<br />

functional or anatomical<br />

asplenia’.<br />

Check the person’s<br />

vaccination status<br />

for pneumococcal,<br />

meningococcal, influenza<br />

and Hib vaccinations.<br />

Persons with a severe or<br />

chronic illness may be at<br />

increased risk of vaccinepreventable<br />

diseases (e.g.<br />

invasive pneumococcal<br />

disease), but may not<br />

mount an optimal immune<br />

response to certain<br />

vaccines.<br />

<strong>The</strong> safety and<br />

effectiveness of some<br />

vaccines may be<br />

suboptimal in persons who<br />

are immunocompromised<br />

(see above).<br />

Intramuscular injection<br />

may lead to haematomas<br />

in patients with disorders<br />

of haemostasis.<br />

Some Indigenous persons<br />

are at increased risk of<br />

some vaccine-preventable<br />

diseases, such as influenza,<br />

pneumococcal disease and<br />

hepatitis A.<br />

Persons with an absent<br />

or dysfunctional spleen<br />

are at an increased risk of<br />

severe bacterial infections,<br />

most notably invasive<br />

pneumococcal disease.<br />

PART 2 VACCINATION PROCEDURES 35<br />

2.1 PRE-VACCINATION


Condition or<br />

circumstance of person<br />

to be vaccinated<br />

Is a parent, grandparent<br />

or carer of an infant<br />

≤6 months of age<br />

Lives with someone who<br />

is immunocompromised<br />

Action Rationale 13-15<br />

Ensure parents,<br />

grandparents and carers of<br />

infants up to 6 months of<br />

age have been offered all<br />

vaccines recommended for<br />

their age group, including<br />

dTpa. ‡<br />

Ensure all recommended<br />

vaccines (in particular<br />

MMR, varicella and<br />

influenza vaccines)<br />

have been offered to<br />

household members of<br />

immunocompromised<br />

persons.<br />

See above and<br />

3.3.3 Vaccination of<br />

immunocompromised<br />

persons.<br />

Is planning travel See 3.2 Vaccination for<br />

international travel<br />

Has certain occupation<br />

or lifestyle factors<br />

See 3.3 Groups with special<br />

vaccination requirements,<br />

and ‘Catch-up schedules<br />

for persons ≥10 years<br />

of age’ in 2.1.5 Catch-up<br />

below.<br />

* Live attenuated vaccines are classified in Table 2.1.3 below.<br />

† See 4.9 Measles, 4.11 Mumps or 4.18 Rubella for further information.<br />

‡ See 4.2 Diphtheria, 4.12 Pertussis or 4.19 Tetanus for further information.<br />

36 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Persons in close contact<br />

are the most likely sources<br />

of vaccine-preventable<br />

diseases, in particular<br />

pertussis, in the newborn.<br />

Household members are<br />

the most likely sources<br />

of vaccine-preventable<br />

diseases among<br />

immunocompromised<br />

persons (who often are<br />

unable to be vaccinated,<br />

especially with live viral<br />

vaccines).<br />

Travellers may be at<br />

increased risk of certain<br />

vaccine-preventable<br />

diseases.<br />

Workers in certain<br />

occupations (e.g.<br />

healthcare workers and<br />

persons working in early<br />

childhood education and<br />

care), and those with<br />

certain lifestyle factors (e.g.<br />

persons who inject drugs)<br />

may be at increased risk of<br />

certain vaccine-preventable<br />

diseases.


Table 2.1.3: Live attenuated parenteral and oral vaccines<br />

Live attenuated parenteral vaccines Live attenuated oral vaccines<br />

Viral Bacterial Viral Bacterial<br />

Japanese encephalitis<br />

(Imojev)<br />

Measles-mumpsrubella<br />

(MMR)<br />

Measles-mumpsrubella-varicella<br />

(MMRV)<br />

Varicella<br />

Yellow fever<br />

Zoster<br />

BCG Oral rotavirus<br />

vaccine<br />

Oral typhoid<br />

vaccine<br />

Contraindications to vaccination<br />

<strong>The</strong>re are only two absolute contraindications applicable to all vaccines:<br />

• anaphylaxis following a previous dose of the relevant vaccine<br />

• anaphylaxis following any component of the relevant vaccine.<br />

<strong>The</strong>re are two further contraindications applicable to live (both parenteral and<br />

oral) vaccines:<br />

• Live vaccines (see Table 2.1.3) should not be administered to persons<br />

who are significantly immunocompromised, regardless of whether the<br />

immunocompromise is caused by disease or treatment. <strong>The</strong> exception<br />

is that, with further advice, MMR, varicella and zoster vaccines can be<br />

administered to HIV-infected persons in whom immunocompromise is<br />

mild. (See 3.3.3 Vaccination of immunocompromised persons, and individual<br />

disease-specific chapters.)<br />

• In general, live vaccines should not be administered during pregnancy,<br />

and women should be advised not to become pregnant within 28 days of<br />

receiving a live vaccine (see Table 3.3.1 Recommendations for vaccination in<br />

pregnancy in 3.3 Groups with special vaccination requirements).<br />

False contraindications to vaccination<br />

No-one should be denied the benefits of vaccination by withholding<br />

vaccines for inappropriate reasons.<br />

Conditions listed in Table 2.1.4 below are not contraindications to<br />

vaccination. Persons with these conditions should be vaccinated with all<br />

recommended vaccines.<br />

PART 2 VACCINATION PROCEDURES 37<br />

2.1 PRE-VACCINATION


Table 2.1.4: False contraindications to vaccination<br />

<strong>The</strong> following conditions are not contraindications to any of the vaccines in the<br />

National <strong>Immunisation</strong> Program schedule:<br />

• mild illness without fever (T


An online ‘catch-up calculator’ for NIP vaccines is hosted by South Australia<br />

Health (at immunisationcalculator.sahealth.sa.gov.au) and is available to assist in<br />

determining appropriate catch-up schedules for children ≤7 years of age across<br />

Australia. When using such resources, also check the accuracy of information<br />

provided by referring to your current state/territory immunisation schedule and<br />

the current edition of the <strong>Handbook</strong>.<br />

If still uncertain about how to plan the catch-up schedule, or for more<br />

complicated catch-up scenarios, seek further advice (see Appendix 1 Contact<br />

details for <strong>Australian</strong>, state and territory government health authorities and<br />

communicable disease control).<br />

For recently arrived migrants, the World Health Organization website (www.<br />

who.int/countries/en) lists immunisation schedules provided by other countries,<br />

which may supplement information regarding which vaccines a child/adult<br />

arriving from overseas may have received (see also 3.3.8 Vaccination of migrants<br />

to Australia).<br />

Confirmation of vaccination history<br />

<strong>The</strong> most important requirement for assessment of vaccination status is to have<br />

written documentation of vaccination. <strong>The</strong> approach of immunisation service<br />

providers to the problem of inadequate records should be based on the age of the<br />

person to be vaccinated, whether previous vaccines have been given in Australia<br />

or overseas, and the vaccines being considered for catch-up.<br />

Detailed information on the vaccine registers used in Australia and how to<br />

obtain vaccination records is provided in 2.3.4 <strong>Immunisation</strong> registers, but is also<br />

described briefly below.<br />

Children


a person who is now ≥7 years of age can be made available to an immunisation<br />

service provider or parent/carer.<br />

<strong>The</strong> National HPV Vaccination Program Register (NHVPR, also referred to as<br />

the ‘HPV Register’) holds details of human papillomavirus (HPV) vaccinations<br />

reported to the Register since the commencement of the HPV Vaccination<br />

Program in April 2007. <strong>The</strong> NHVPR initially only recorded vaccinations for<br />

females, but from <strong>2013</strong> will also record vaccinations given to males. Details of<br />

HPV vaccinations held by the NHVPR can be obtained by phoning the Register<br />

on 1800 478 734 (1800 HPV REG). (See also 2.3.4 <strong>Immunisation</strong> registers.)<br />

In older children and adolescents, alternative sources of documentation (such as<br />

personal health records) will be needed, but are less likely to be available with<br />

increasing age. Persons who do not have personal vaccination records may seek<br />

evidence of past vaccination from their parents, their past and present healthcare<br />

providers or immunisation service providers. Those born after 1990 may have<br />

some vaccinations recorded on the ACIR (see 2.3.4 <strong>Immunisation</strong> registers).<br />

Information on how to obtain records of vaccines received through school-based<br />

vaccination programs can be obtained from state and territory government health<br />

departments (see Appendix 1 Contact details for <strong>Australian</strong>, state and territory<br />

government health authorities and communicable disease control).<br />

Adults (≥18 years of age)<br />

In adults, patient-held and/or provider-held documentation of previous<br />

vaccination history may not be available. In some cases, information may be<br />

available from other sources, such as the National HPV Vaccination Program<br />

Register and the <strong>Australian</strong> Q Fever Register. (See also 2.3.4 <strong>Immunisation</strong><br />

registers.)<br />

Incomplete documentation of prior vaccination<br />

If receipt of prior vaccination cannot be confirmed via the above methods, it<br />

should generally be assumed that the vaccine(s) required have not been given<br />

previously. All efforts should be made to confirm and ensure appropriate<br />

documentation of prior receipt of vaccines.<br />

For most vaccines (except Q fever), there are no adverse events associated with<br />

additional doses if given to an already immune person. In the case of diphtheria-,<br />

tetanus- and pertussis-containing vaccines and pneumococcal polysaccharide<br />

vaccines, frequent additional doses may be associated with an increase in local<br />

adverse events; however, the benefits of protection may outweigh the risk of an<br />

adverse reaction, for example, protection against pertussis from a booster dose<br />

of dTpa. (See also 4.2 Diphtheria, 4.12 Pertussis, 4.13 Pneumococcal disease or 4.19<br />

Tetanus.) Additional doses of MMR, varicella, inactivated poliomyelitis (IPV) or<br />

hepatitis B vaccines are rarely associated with significant adverse events.<br />

40 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Use of serological testing to guide catch-up vaccination<br />

In some instances, serological testing for immunity from prior vaccination and/<br />

or infection may be useful to guide the need for catch-up vaccination, such<br />

as for measles, hepatitis B and rubella. However, it is important to note that<br />

serological testing is not reliable for vaccine-induced immunity in all instances<br />

and is specifically not recommended to be used to guide the need for catch-up<br />

vaccination for certain diseases/vaccines (e.g. pertussis, pneumococcal disease<br />

and meningococcal disease). In most circumstances, and for most vaccines, it<br />

is more practical to offer vaccination, rather than serological testing. See also<br />

recommendations regarding serological testing before and after vaccination in<br />

various disease chapters (4.4 Hepatitis A, 4.5 Hepatitis B, 4.9 Measles, 4.11 Mumps,<br />

4.18 Rubella, 4.22 Varicella, 4.24 Zoster).<br />

Interpretation of the results of serological testing may be enhanced by discussion<br />

with the laboratory that performed the test, ensuring that relevant clinical<br />

information is provided.<br />

Determining when a vaccine dose is valid according to age and interval<br />

since last dose<br />

A ‘valid’ vaccine dose is a dose that is considered immunogenic (and safe) given<br />

the age and health status of the recipient and the interval since the recipient’s<br />

last dose of the same vaccine. For children who are vaccinated at an age younger<br />

than that routinely recommended, or for children and adults in whom the<br />

interval between vaccine doses is shorter than the usual recommended interval,<br />

information regarding both the minimum acceptable age for the 1st dose of<br />

an infant vaccine (Table 2.1.5) and the minimum acceptable intervals between<br />

vaccine doses (Tables 2.1.7 to 2.1.12) can be used to determine whether additional<br />

vaccine doses and/or catch-up vaccination is required. For more details see the<br />

following sections.<br />

Planning catch-up vaccination<br />

This and the following two sections are dedicated to planning catch-up<br />

vaccination. In the following two sections information is presented by age<br />

of the vaccine recipient (children aged


• Table 2.1.6 can be used to assess the number of doses a child should have<br />

received if they were on schedule. Check under the current age of the child<br />

to see how many doses they should have already received and use that<br />

number of doses as the starting point for calculating a catch-up schedule.<br />

For example, a child who is 18 months old now should have received<br />

3 doses of DTPa, 3 doses of IPV, etc.<br />

• Table 2.1.7 lists the minimum acceptable interval between doses under special<br />

circumstances, such as catch-up vaccination. Vaccine doses should not be<br />

administered at less than the acceptable minimum interval. 16 In the majority<br />

of instances, doses administered earlier than the minimum acceptable<br />

interval should not be considered as valid doses and should be repeated,<br />

as appropriate, using Table 2.1.6.<br />

• Tables 2.1.8 to 2.1.11 are for calculating catch-up for Haemophilus influenzae<br />

type b (Hib) and pneumococcal vaccination of children.<br />

• Table 2.1.12 can be used to calculate a catch-up schedule for persons<br />

aged ≥10 years.<br />

In addition, the following principles should generally be applied when planning<br />

catch-up vaccination:<br />

• When commencing the catch-up schedule, the standard scheduled interval<br />

between doses may be reduced or extended, and the numbers of doses<br />

required may reduce with age. For example, from 15 months of age, only<br />

1 dose of (any) Hib vaccine is required.<br />

• As a child gets older, the recommended number of vaccine doses may change<br />

(or even be omitted from the schedule), as the child becomes less vulnerable<br />

to specific diseases.<br />

• For incomplete or overdue vaccinations, build on the previous documented<br />

doses. In almost every circumstance, it is advisable to not start the schedule<br />

again, regardless of the interval since the last dose, but to count previous<br />

doses. One exception to this rule is for oral cholera vaccine (see 4.1 Cholera).<br />

• If more than one vaccine is overdue, 1 dose of each due or overdue vaccine<br />

should be given at the first catch-up visit. Further required doses should be<br />

scheduled after the appropriate minimum interval (see Table 2.1.7).<br />

• A catch-up schedule may require multiple vaccinations at a visit. Give all the<br />

due vaccines at the same visit – do not defer. See 2.2.9 Administering multiple<br />

vaccine injections at the same visit.<br />

• <strong>The</strong> standard intervals and ages recommended in the NIP schedule should be<br />

used once the child or adult is up to date with the schedule.<br />

• Some persons will require further doses of antigens that are available only<br />

in combination vaccines. In general, the use of the combination vaccine(s)<br />

is acceptable, even if this means the number of doses of another antigen<br />

administered exceeds the required number.<br />

42 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


• For some vaccines, catch-up vaccination is not recommended. For<br />

example, rotavirus vaccination is not recommended if the 1st (and<br />

subsequent) vaccine doses are not able to be provided within the<br />

prescribed upper age limits (see ‘Catch-up guidelines for individual<br />

vaccines for children


5. If any variations to the schedule are necessary due to recorded factors (e.g. a<br />

child who is immunocompromised may require different vaccines), adjust the<br />

‘number of doses required’ accordingly.<br />

6. For each vaccine, compare the number of doses received, as recorded in the<br />

‘Last dose given’ column, with the number of doses required for the child’s<br />

current age.<br />

7. If the child has already received the number of doses required for a<br />

particular vaccine, cross through the relevant ‘Dose number due now’ and<br />

‘Further doses’ columns. Ensure that the minimum acceptable interval has<br />

been observed for all doses previously received, particularly if the child<br />

commenced their vaccination program overseas.<br />

8. If the number of doses received, as recorded in the ‘Last dose given’ column,<br />

is less than the number of doses required, administer a dose of the relevant<br />

vaccine now, and record this in the ‘Dose number due now’ column. If<br />

this dose still does not complete the required doses, enter the further dose<br />

numbers in the ‘Further doses’ column.<br />

9. To schedule the next dose at the most appropriate time (usually at the<br />

earliest opportunity), refer to Table 2.1.7 for the minimum acceptable interval<br />

required between doses. Record when the next dose is due in the ‘Further<br />

doses’ column.<br />

10. Convert this information into a list of proposed appointment dates,<br />

detailing vaccines and dose number needed at each visit on the ‘Catch-up<br />

appointments’ section of the worksheet.<br />

11. Record this catch-up schedule in your provider records and provide a copy to<br />

the child’s parent/carer.<br />

12. Once a child has received relevant catch-up vaccines, give the remaining<br />

scheduled vaccines as per the recommended NIP schedule. For example, for a<br />

12-month-old child who is brought up to date with all vaccines including the<br />

12-month vaccinations, the 2nd dose of MMR-containing vaccine should be<br />

given at 18 months of age, not 4 weeks after the last received dose.<br />

44 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Figure 2.1.1: Catch-up worksheet for children


Table 2.1.5: Minimum acceptable age for the 1st dose of scheduled vaccines in<br />

infants in special circumstances*<br />

Vaccine Minimum age<br />

for 1st dose<br />

in special<br />

circumstances*<br />

46 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Action if a vaccine dose is inadvertently<br />

administered prior to the recommended<br />

minimum age 17<br />

DTPa 6 weeks If the 1st dose of DTPa-containing vaccine<br />

was administered at ≤28 days of age, it is<br />

recommended that the dose is repeated. This<br />

repeat dose should be given at 2 months of age.<br />

<strong>The</strong> NIP schedule should be followed thereafter,<br />

with the next dose of DTPa-containing vaccine<br />

given at 4 months of age. †‡<br />

Poliomyelitis<br />

(IPV)<br />

If the 1st dose of DTPa-containing vaccine<br />

was administered between >28 days and<br />

28 days and


Vaccine Minimum age<br />

for 1st dose<br />

in special<br />

circumstances*<br />

Action if a vaccine dose is inadvertently<br />

administered prior to the recommended<br />

minimum age 17<br />

Rotavirus 6 weeks If the 1st dose of rotavirus vaccine was<br />

administered at ≤28 days of age, it is<br />

recommended that the dose is repeated.<br />

This repeat dose should be given at 2 months<br />

of age. <strong>The</strong> NIP schedule should be followed<br />

thereafter, with the next dose of rotavirus vaccine<br />

given at 4 months of age.<br />

If the 1st dose of rotavirus vaccine was<br />

administered between >28 days and


* Special circumstances may include infants/children being vaccinated during an outbreak of a<br />

certain disease, before overseas travel, or opportunistic vaccination following early attendance<br />

to a provider. <strong>The</strong>se ages will often differ from routinely recommended ages of administration<br />

under the NIP schedule. In some instances, these ages will also result in the dose not being<br />

considered by the <strong>Australian</strong> Childhood <strong>Immunisation</strong> Register (ACIR) as ‘valid’ for the purpose<br />

of calculating immunisation status. If the ACIR age requirement differs from the minimum ages<br />

in this table, this is noted.<br />

† If the need to repeat the 1st dose of vaccine is not recognised until the infant is older (e.g. a<br />

4-month-old infant presents for vaccination and has only previously received 1 dose of DTPahepB-IPV-Hib<br />

or 13vPCV vaccines both at age 28 days) of age. 17<br />

§ Monovalent hepatitis B vaccine should be given at birth (up to 7 days of age). However, for<br />

subsequent doses where hepatitis B-containing combination vaccine is given at 2, 4 and 6 months<br />

of age, the minimum age for the 1st dose (scheduled at age 2 months) is 6 weeks of age. If an<br />

infant has not received a birth dose within the first 7 days of life, a primary 3-dose course of a<br />

hepatitis B-containing combination vaccine should be given at 2, 4 and 6 months of age; catch-up<br />

of the birth dose is not necessary.<br />

MMRV vaccine is recommended as the 2nd (not 1st) dose of MMR-containing vaccine in<br />

children


Table 2.1.6: Number of vaccine doses that should have been administered by<br />

the current age of the child<br />

This table can be used in conjunction with Figure 2.1.1 Catch-up worksheet for children<br />


Table 2.1.7: Minimum acceptable dose intervals for children


Catch-up guidelines for individual vaccines for children


MMR vaccine, MMRV vaccine and varicella vaccine<br />

If no previous documented doses have been given, catch-up for MMR vaccine<br />

consists of 2 doses of MMR-containing vaccine, given at least 4 weeks apart<br />

(see 4.9 Measles). If no previous documented varicella vaccination has been<br />

given, a single dose of varicella-containing vaccine is recommended in children<br />

aged


If 13vPCV is not available, and 10vPCV is being used, 10vPCV is recommended<br />

in a 4-dose schedule. (See also 4.13 Pneumococcal disease.) If catch-up is required<br />

for 10vPCV, vaccination can be done according to the information provided in<br />

Table 2.1.10.<br />

Children aged ≥5 years who are not at increased risk of invasive pneumococcal<br />

disease (including Indigenous children aged ≥5 years) do not require catch-up<br />

doses of PCV.<br />

Preterm infants born at


Table 2.1.8: Catch-up schedule for Haemophilus influenzae type b (Hib)<br />

vaccination for children


Previous<br />

vaccination<br />

history<br />

2 previous<br />

doses of<br />

PRP-T<br />

(or<br />

1 of each of<br />

PRP-OMP and<br />

PRP-T)<br />

3 previous<br />

doses<br />

(all PRP-T or<br />

at least 1 PRP-<br />

OMP)<br />

Age at Type<br />

presentation of Hib<br />

vaccine to<br />

be used †<br />

7–11 months PRP-OMP<br />

or PRP-T<br />

12–59 months PRP-OMP<br />

or PRP-T<br />

7–11 months PRP-OMP<br />

or PRP-T<br />

12–59 months PRP-OMP<br />

or PRP-T<br />

1st<br />

dose<br />

Previously<br />

given<br />

Previously<br />

given<br />

Previously<br />

given<br />

Previously<br />

given<br />

2nd<br />

dose<br />

Previously<br />

given<br />

Previously<br />

given at<br />

≤15 months<br />

of age ‡<br />

Previously<br />

given<br />

Previously<br />

given<br />

3rd<br />

dose<br />

Give<br />

now §<br />

Not<br />

needed<br />

Previously<br />

given<br />

Previously<br />

given at<br />

≤15 months<br />

of age ‡<br />

Booster<br />

dose<br />

12 months<br />

of age or<br />

2 months<br />

after 2nd dose<br />

(whichever is<br />

later)<br />

Give now ‡<br />

or 2 months<br />

after 2nd dose<br />

(whichever is<br />

later)<br />

12 months<br />

of age or<br />

2 months<br />

after 2nd dose<br />

(whichever is<br />

later)<br />

Give now ‡<br />

or 2 months<br />

after 2nd dose<br />

(whichever is<br />

later)<br />

* Recommendations for vaccination of haematopoietic stem cell transplant (HSCT) recipients<br />

differ; see Table 3.3.3 Recommendations for revaccination following HSCT in children and adults,<br />

irrespective of previous immunisation history.<br />

† PRP-OMP is the Hib formulation contained in Liquid PedvaxHIB. PRP-T is the Hib formulation<br />

contained in the other Hib-containing vaccines: ACT-Hib, Hiberix, Infanrix hexa, Menitorix<br />

and Pediacel. A minimum interval of 1 month is recommended between primary doses of Hibcontaining<br />

vaccine in a catch-up schedule.<br />

‡ A booster dose is not needed if the last previous dose was given at >15 months of age.<br />

§ If the 1st dose of Hib vaccine was given at >6 months of age, a 3rd dose, prior to the booster<br />

dose at age ≥12 months, is not required.<br />

PART 2 VACCINATION PROCEDURES 55<br />

2.1 PRE-VACCINATION


56 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Table 2.1.9: Catch-up schedule for 13vPCV (Prevenar 13) for non-Indigenous children, and Indigenous children residing<br />

in the <strong>Australian</strong> Capital Territory, New South Wales, Tasmania and Victoria, who do not have any medical<br />

condition(s) associated with an increased risk of invasive pneumococcal disease (IPD), aged


PART 2 VACCINATION PROCEDURES 57<br />

Table 2.1.10: Catch-up schedule for 13vPCV* (Prevenar 13) for Indigenous children residing in the Northern Territory,<br />

Queensland, South Australia or Western Australia ONLY, who do not have any medical condition(s) associated<br />

with an increased risk of invasive pneumococcal disease (IPD), aged


58 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Number of doses<br />

given previously<br />

2 previous doses<br />

3 previous doses<br />

Age at<br />

presentation<br />


PART 2 VACCINATION PROCEDURES 59<br />

Table 2.1.11: Catch-up schedule for 13vPCV (Prevenar 13) and 23vPPV (Pneumovax 23) in children with a medical condition(s)<br />

associated with an increased risk of invasive pneumococcal disease (IPD),* presenting at age


60 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Number of<br />

doses given<br />

previously<br />

2 previous<br />

doses<br />

3 previous<br />

doses<br />

Age at<br />

presentation<br />

Age when previous dose of any PCV ‡ was given Recommendations<br />

1st dose 2nd dose 3rd dose Number<br />

of further<br />

primary dose(s)<br />

of 13vPCV<br />

required §<br />

Number of<br />

booster doses<br />

of 13vPCV<br />

required at<br />

age ≥12months <br />

Number<br />

of doses<br />

of 23vPPV<br />

required at<br />

age 4–5 years<br />


Catch-up schedules for persons ≥10 years of age<br />

Catch-up is much less commonly required for this age group than for young<br />

children. Nevertheless, issues surrounding booster doses or revaccinations are<br />

common, particularly in adults. Persons who did not have natural infection<br />

as children but were not vaccinated remain at unnecessary risk of vaccinepreventable<br />

diseases.<br />

In general, the same principles for catch-up vaccination apply as for younger<br />

children. For example, if a vaccine course is incomplete, do not start the course<br />

again, regardless of the interval since the last dose. One exception to this rule is<br />

for oral cholera vaccine (see 4.1 Cholera).<br />

Catch-up vaccination for adults can be less straightforward than for children and<br />

adolescents. A useful principle to consider when planning which vaccines to give<br />

to adults is the HALO principle, which allows for assessment of vaccines needed<br />

depending on risk factors:<br />

• Health<br />

• Age<br />

• Lifestyle<br />

• Occupation<br />

<strong>The</strong> schedule for each individual adult may differ because of the risk factors<br />

identified when applying the HALO principle. Some examples of how the HALO<br />

principle can be used:<br />

• Health: the person to be vaccinated has a medical condition(s) that places<br />

them at increased risk of acquiring a particular vaccine-preventable disease<br />

or experiencing complications from that disease, for example, influenza.<br />

• Age: older age groups may require extra vaccines, such as influenza or<br />

pneumococcal vaccination, or certain age groups may be targeted for<br />

immunisation against a particular vaccine-preventable disease, such as HPV.<br />

Another example is young to middle-aged adults who may have missed out<br />

on vaccine doses due to schedule changes, such as the 2nd dose of MMR<br />

vaccine.<br />

• Lifestyle: the person may have missed vaccines because they moved location<br />

of residence, may require extra vaccines because they travel frequently, or<br />

have other lifestyle risk factors that increase their risk of acquiring a vaccinepreventable<br />

disease, for example, smoking or injecting drugs.<br />

• Occupation: the person may be employed in an occupation for which certain<br />

vaccines are recommended because of the increased risk of acquiring a<br />

vaccine-preventable disease and/or transmitting it to others, such as in<br />

healthcare or early childhood education and care.<br />

<strong>The</strong> HALO principle is also incorporated, to some extent, into questions used in<br />

the pre-vaccination screening checklist (see Tables 2.1.1 and 2.1.2).<br />

PART 2 VACCINATION PROCEDURES 61<br />

2.1 PRE-VACCINATION


Table 2.1.12 contains information on vaccine doses and intervals between doses<br />

for persons aged ≥10 years in whom catch-up vaccination is required. This table<br />

only contains information on vaccines that are recommended at a population<br />

level, and for which catch-up is required if doses have been missed earlier in<br />

life. <strong>The</strong> table does not include information on all vaccines required for adults.<br />

Recommended vaccines and catch-up vaccination that might be required when<br />

assessed using the HALO principle above and/or by using the pre-vaccination<br />

screening checklist (see Tables 2.1.1 and 2.1.2) are discussed in 3.3 Groups with<br />

special vaccination requirements.<br />

Table 2.1.12 can be used as follows:<br />

• determine how many doses of a particular vaccine a person should have<br />

received to be considered completely vaccinated (see ‘Doses required’<br />

column)<br />

• deduct any previous doses of the vaccine from the number in the ‘Doses<br />

required’ column<br />

• check the appropriate ‘Minimum interval’ column to schedule further doses<br />

• refer to the relevant disease-specific chapter(s) in Part 4, 3.1 Vaccination for<br />

Aboriginal and Torres Strait Islander people, 3.2 Vaccination for international<br />

travel, and 3.3 Groups with special vaccination requirements, for additional<br />

recommendations, as required.<br />

For example, a 32-year-old woman (Age) is returning to nursing (Occupation)<br />

but has only ever had 1 dose of hepatitis B vaccine, 4 doses of the oral<br />

poliomyelitis vaccine, 1 dose of MMR vaccine and 2 doses of DTPw vaccine as a<br />

child and recently had a splenectomy (Health) following an accident. This person<br />

would require:<br />

• 1 dose of dTpa<br />

• 2 adult doses of hepatitis B; 1 dose given now and a further dose in 2 months<br />

• no further doses of poliomyelitis vaccine (is fully vaccinated against<br />

poliomyelitis)<br />

• 1 dose of MMR vaccine<br />

• 2 doses of varicella vaccine if non-immune; 1 dose given now and a further<br />

dose in 4 weeks<br />

• 1 dose of influenza vaccine, and 1 dose annually thereafter<br />

• pneumococcal vaccine: 1 dose of 13vPCV, followed by 23vPPV approximately<br />

2 months later (because of splenectomy)<br />

• 1 dose of Hib vaccine (because of splenectomy)<br />

• 2 doses of 4vMenCV; 1 dose given now and a further dose in 8 weeks<br />

(because of splenectomy).<br />

62 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


For additional details on these recommendations, see 3.3.7 Vaccination of persons<br />

at occupational risk; ‘Persons with functional or anatomical asplenia’ in 3.3.3<br />

Vaccination of immunocompromised persons, and relevant disease-specific chapters<br />

in Part 4.<br />

Where several vaccines are required for an adolescent or adult – for example,<br />

dTpa, hepatitis B and poliomyelitis vaccines – childhood combination vaccines<br />

(recommended for use in those


Vaccine Doses<br />

required<br />

Zoster vaccine 1 dose if aged<br />

≥60 years<br />

64 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Minimum<br />

interval<br />

between<br />

dose 1 and 2<br />

Minimum<br />

interval<br />

between<br />

dose 2 and 3<br />

Not required Not required<br />

* One of the doses should be given as dTpa (or dTpa-IPV if poliomyelitis vaccination is also<br />

needed) and the course completed with dT. In the unlikely event that dT is not available, dTpa or<br />

dTpa-IPV may be used for all 3 primary doses but this is not routinely recommended as there are no<br />

data on the safety, immunogenicity or efficacy of dTpa for primary vaccination (see also 4.12 Pertussis).<br />

† For hepatitis B vaccine, the minimum interval between dose 1 and dose 3 is 4 months (see 4.5<br />

Hepatitis B).<br />

‡ 4vMenCV is indicated for those at increased risk of meningococcal disease; see recommendations<br />

in 4.10 Meningococcal disease and 3.3 Groups with special vaccination requirements).<br />

§ Varicella vaccine is recommended for all non-immune persons. At least 1 dose should be given<br />

to those aged


2.2 ADMINISTRATION OF VACCINES<br />

2.2.1 Occupational health and safety issues<br />

Standard occupational health and safety guidelines should always be followed<br />

during a vaccination. This will minimise the risk of needle-stick injury. 1<br />

Work practices should include the use of standard infection control precautions<br />

to minimise exposure to blood and body fluids (refer to National Health and<br />

Medical Research Council’s <strong>Australian</strong> guidelines for the prevention and control<br />

of infection in healthcare). 1 If exposure does occur, guidelines for post-exposure<br />

prophylaxis should be followed. Gloves are not routinely recommended for<br />

immunisation service providers, unless the person administering the vaccine is<br />

likely to come into contact with body fluids or has open lesions on the hands.<br />

A new, sterile, disposable syringe and needle must be used for each injection.<br />

Disposable needles and syringes must be discarded into a clearly labelled,<br />

puncture-proof, spill-proof container that meets <strong>Australian</strong> standards in order to<br />

prevent needle-stick injury or re-use. 1 Always keep sharps containers out of the<br />

reach of children. All immunisation service providers should be familiar with the<br />

handling and disposal of sharps according to the National Health and Medical<br />

Research Council’s <strong>Australian</strong> guidelines for the prevention and control of infection in<br />

healthcare. 1<br />

2.2.2 Equipment for vaccination<br />

Preparing for vaccination<br />

Depending on the vaccine(s) that are to be administered, and the age and size of<br />

the person to be vaccinated, decide on the appropriate injection site and route,<br />

and the injection equipment required (e.g. syringe size, needle length and gauge).<br />

<strong>The</strong> equipment chosen will vary depending on whether the vaccine is a<br />

reconstituted vaccine, a vaccine from an ampoule or vial, or a vaccine in a prefilled<br />

syringe.<br />

Equipment may include:<br />

• medical waste (sharps) container<br />

• vaccine, plus diluent if reconstitution is required<br />

• 2 or 3 mL syringe (unless vaccine is in pre-filled syringe)<br />

• appropriate drawing-up needle (19 or 21 gauge needle if required, to draw<br />

up through rubber bung and for reconstitution of vaccine)<br />

• appropriate injecting needle (see Table 2.2.2 Recommended needle size, length<br />

and angle for administering vaccines)<br />

• clean cotton wool and hypoallergenic tape to apply to injection site after<br />

vaccination<br />

• a rattle or noisy toy for distraction after the injection.<br />

PART 2 VACCINATION PROCEDURES 65<br />

2.2 ADMINISTRATION<br />

OF VACCINES


Preparing the vaccine<br />

• Ensure that the minimum/maximum thermometer displays temperatures<br />

within the +2°C to +8°C range before removing vaccine from the refrigerator.<br />

• Ensure that the correct vaccine is taken from the refrigerator and that it is<br />

within the expiry date.<br />

• Check that there is no particulate matter or colour change in the vaccine.<br />

• Ensure that the diluent container is not damaged and potentially<br />

contaminated.<br />

• Wash hands with soap and water or use a waterless alcohol-based hand rub. 2<br />

• Prepare the appropriate injection equipment for the vaccine to be<br />

administered.<br />

Injectable vaccines that do not require reconstitution<br />

• If the vaccine is in a vial, remove the cap carefully to maintain sterility of the<br />

rubber bung. <strong>The</strong>re is no need to wipe the rubber bung of single-dose vials<br />

with an alcohol swab if it is visibly clean. If there is visible contamination, the<br />

bung should be cleaned with a single-use swab, allowing time to dry before<br />

drawing up the contents. 3<br />

• Use a 19 or 21 gauge needle to draw up the recommended dose through the<br />

bung (or through the top of the ampoule), if required.<br />

• Change the needle after drawing up from a vial with a rubber bung or<br />

ampoule, before giving the injection. If using a safety needle system, once<br />

the vaccine has been drawn up, draw back on the syringe to ensure as much<br />

vaccine as possible is removed from the tip of the needle, and then eliminate<br />

any air to the tip of the syringe without re-priming the needle.<br />

Injectable vaccines that require reconstitution<br />

• Reconstitute the vaccine as needed immediately before administration.<br />

• Use a sterile 21 gauge needle for reconstitution and a separate 23 or 25<br />

gauge needle, 25 mm in length, for administration of the vaccine in most<br />

circumstances.<br />

• Use only the diluent supplied with the vaccine; do not use sterile water for<br />

injection instead of a supplied diluent. Ensure that the diluent and vaccine<br />

are completely mixed. 4<br />

• Check reconstituted vaccines for signs of deterioration, such as a change in<br />

colour or clarity.<br />

• Administer reconstituted vaccines as soon as practicable after they have been<br />

reconstituted as they may deteriorate rapidly. Refer to individual vaccine<br />

product information for recommended times from vaccine reconstitution to<br />

administration.<br />

66 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


• Never freeze a vaccine after it has been reconstituted.<br />

For all injectable vaccines<br />

• Do not extrude small air bubbles through the needle for injection. However,<br />

in the rare instance of a large air bubble in a pre-filled syringe, first draw back<br />

on the needle to ensure no vaccine is expelled along with the air, and then<br />

expel the air through the needle, taking care not to prime the needle with any<br />

of the vaccine, as this can lead to increased local reaction.<br />

• Never mix other vaccines together in the one syringe (unless that is the<br />

manufacturer’s registered recommendation, e.g. Infanrix hexa). 4<br />

• Never mix a local anaesthetic with a vaccine. 4<br />

Vaccines in multi-dose vials<br />

Multi-dose vials are not routinely used in Australia. <strong>The</strong> current exception<br />

is bacille Calmette-Guérin (BCG) vaccine (see 4.20 Tuberculosis). Single-dose<br />

preparations are now available for all other vaccines currently on the NIP.<br />

However, where mass vaccination of a population is required, such as during the<br />

2009–2010 H1N1 influenza pandemic, multi-dose vials have some advantages<br />

over single-dose vaccines. <strong>The</strong> production of vaccines in multi-dose vials is more<br />

cost effective and can also mean that the vaccine takes less time to manufacture.<br />

Multi-dose vials also take up less storage room in a vaccine fridge. 5<br />

<strong>The</strong> primary risk with use of multi-dose vials is a breach in infection control<br />

through user error, for example, an unsterile needle is inserted into the vial<br />

or a contaminated syringe is re-used. It is recognised that there are multiple<br />

reports of instances of transmission of bacteria or blood-borne viruses through<br />

inappropriate use of multi-dose vials; however, the majority of these have been in<br />

high-risk settings such as haemodialysis units or with use of anaesthetics and did<br />

not involve immunisations.<br />

2.2.3 Route of administration<br />

Most vaccines available in Australia are given intramuscularly. Only a few<br />

vaccines are given subcutaneously, orally or intradermally.<br />

Rotavirus vaccines are only available for oral administration and must never<br />

be injected.<br />

Special training is required for intradermal administration, which is important<br />

for several vaccines (see 4.15 Q fever and 4.20 Tuberculosis).<br />

Table 2.2.1 summarises the route of administration for vaccines used in Australia.<br />

PART 2 VACCINATION PROCEDURES 67<br />

2.2 ADMINISTRATION<br />

OF VACCINES


68 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Table 2.2.1: Route of administration for vaccines used in Australia<br />

Intramuscular (IM) injection Subcutaneous (SC) injection IM or SC injection Intradermal Oral<br />

Diphtheria-tetanus vaccine (dT) Inactivated poliomyelitis<br />

Diphtheria-tetanus-acellular<br />

vaccine (IPV)*<br />

pertussis vaccine (DTPa and<br />

dTpa)<br />

DTPa- and dTpa-combination<br />

Quadrivalent meningococcal<br />

polysaccharide vaccine<br />

(4vMenPV)<br />

vaccines<br />

Varicella vaccine (VV)<br />

Hepatitis A vaccine and<br />

Japanese encephalitis vaccine<br />

Hepatitis A combination<br />

(Imojev)<br />

vaccines<br />

Q fever vaccine<br />

Hepatitis B vaccine and Hepatitis<br />

B combination vaccines<br />

Haemophilus influenzae type b<br />

(Hib) vaccine<br />

Human papillomavirus (HPV)<br />

vaccine<br />

IPV-containing combination<br />

vaccines*<br />

Japanese encephalitis vaccine<br />

(JEspect)<br />

10-valent pneumococcal<br />

conjugate vaccine (10vPCV)<br />

13-valent pneumococcal<br />

conjugate vaccine (13vPCV)<br />

Typhoid Vi polysaccharide<br />

vaccine<br />

‡<br />

Influenza vaccine<br />

Measles-mumps-rubella<br />

vaccine (MMR) (M-M-R II<br />

only)<br />

Measles-mumps-rubellavaricella<br />

vaccine (MMRV)<br />

(ProQuad only)<br />

Zoster vaccine<br />

†<br />

Measles-mumps- rubella<br />

vaccine (MMR) (Priorix only)<br />

Measles-mumps-rubellavaricella<br />

vaccine (MMRV)<br />

(Priorix-tetra only)<br />

23-valent pneumococcal<br />

polysaccharide vaccine<br />

(23vPPV) †<br />

Influenza vaccine<br />

(Intanza only)<br />

Bacille Calmette-Guérin<br />

(BCG) vaccine<br />

Rabies vaccine (HDCV)<br />

Yellow fever vaccine<br />

‡<br />

Q fever skin testing ‡<br />

Rotavirus vaccine<br />

Cholera vaccine<br />

Typhoid vaccine


PART 2 VACCINATION PROCEDURES 69<br />

Intramuscular (IM) injection Subcutaneous (SC) injection IM or SC injection Intradermal Oral<br />

Meningococcal C conjugate<br />

vaccine (MenCCV)<br />

Quadrivalent meningococcal<br />

conjugate vaccine (4vMenCV)<br />

Rabies vaccine (PCECV)<br />

* IPV-containing combination vaccines are administered by IM injection; IPV (IPOL) is administered by SC injection.<br />

† <strong>The</strong> IM route is preferred to the SC route because it causes fewer local adverse events. 6,7<br />

‡ Q fever skin testing and BCG vaccine should be administered only by specially trained immunisation service providers.<br />

2.2 ADMINISTRATION<br />

OF VACCINES


2.2.4 Preparation for vaccine administration<br />

Skin cleaning<br />

Provided the skin is visibly clean, there is no need to wipe it with an antiseptic<br />

(e.g. alcohol wipe). 3,8 If the immunisation service provider decides to clean the<br />

skin, or if the skin is visibly not clean, alcohol and other disinfecting agents must<br />

be allowed to dry before vaccine injection (to prevent inactivation of live vaccines<br />

and to reduce the likelihood of irritation at the injection site). 9<br />

Distraction techniques<br />

<strong>The</strong> routine use of distraction, relaxation and other measures have been shown to<br />

reduce distress and pain following vaccination in young children. 10-13 Reducing<br />

children’s distress may enhance parents’ timely attendance for subsequent<br />

vaccinations.<br />

Distraction measures that may decrease discomfort following vaccination in<br />

young children include: 10-13<br />

• swaddling and holding the infant securely (but not excessively)<br />

• shaking a noisy toy (for infants and very young children)<br />

• playing music<br />

• encouraging an older child to pretend to blow away the pain using a<br />

windmill toy or bubbles<br />

• breastfeeding the infant during administration of the vaccine.<br />

Discomfort may also be decreased by administering sweet-tasting fluid orally<br />

immediately before the injection (with parental consent). In infants, 15–25%<br />

sucrose drops have been used. 14<br />

Topical anaesthetic agents, including vapocoolant sprays, are available but, to<br />

be effective, must be applied at the correct time before vaccine administration.<br />

Topical anaesthetics, such as EMLA, are not recommended for routine use, but<br />

could be considered in a child with excessive fear or dislike of needles; they<br />

require application 30 to 60 minutes before an injection. 15<br />

Vapocoolant sprays are applied 15 seconds before vaccination. <strong>The</strong>se sprays have<br />

been shown to be more effective in adults than children as children can perceive<br />

coldness as painful and spray application may also focus the child more on the<br />

procedure. Topical lignocaine/prilocaine is not recommended for children<br />


2.2.5 Vaccine injection techniques<br />

Intramuscular injection technique 16,17<br />

• For intramuscular (IM) injection, use a 25 mm needle in most cases<br />

(see Table 2.2.2).<br />

• Depending on the injection site, position the limb so as to relax the muscle<br />

into which the vaccine is to be injected.<br />

• Pierce the skin at an angle of 90° to the skin, so the needle can be safely<br />

inserted to the hub. 18 Provided an injection angle of >70° is used, the needle<br />

should reach the muscle layer. 19<br />

• If using a 25 gauge needle for an IM vaccination, ensure the vaccine is<br />

injected slowly over a count of 5 seconds to avoid injection pain and muscle<br />

trauma (see Table 2.2.2).<br />

• If you have drawn back on the syringe plunger before injecting a vaccine<br />

(which is not considered necessary), 10 and a flash of blood appears in the<br />

needle hub, withdraw the needle and select a new site for injection. 20<br />

Studies have demonstrated that, for most vaccines, local adverse events<br />

are minimised and immunogenicity is enhanced by ensuring vaccine is<br />

deposited into the muscle and not into the subcutaneous layer. 10,21-24 However,<br />

some vaccines (e.g. inactivated poliomyelitis, varicella and meningococcal<br />

polysaccharide vaccines) are only registered for SC administration (see Table<br />

2.2.1).<br />

In the instance where a vaccine that is registered for administration only via<br />

the IM route is inadvertently administered via the SC route, check the vaccine<br />

product information and the ‘Vaccines’ section in relevant disease-specific<br />

chapters in Part 4 for additional information. Some vaccines may still be<br />

immunogenic when given via the SC route, and as such, would not need to<br />

be repeated. One vaccine that should be considered invalid and that therefore<br />

needs to be repeated is Rabipur Inactivated Rabies Virus Vaccine (PCECV) (see<br />

4.16 Rabies and other lyssaviruses (including <strong>Australian</strong> bat lyssavirus)). In general,<br />

hepatitis B vaccines should also be repeated if inadvertently given SC. However,<br />

in special circumstances, for example, in persons with bleeding disorders, some<br />

hepatitis B vaccines may be given via the SC route (see 3.3.5 Vaccination of persons<br />

with bleeding disorders).<br />

A clinical trial demonstrated that for infant vaccination long (25 mm) needles<br />

(with the skin stretched flat and the needle inserted at 90°) were associated<br />

with significantly fewer local adverse events, while achieving comparable<br />

immunogenicity. Little difference in local adverse events or immune response<br />

was found between needles of the same length but with different gauges. 18<br />

PART 2 VACCINATION PROCEDURES 71<br />

2.2 ADMINISTRATION<br />

OF VACCINES


Subcutaneous injection technique<br />

For subcutaneous (SC) injection, administer the injection at a 45° angle to the<br />

skin. <strong>The</strong> standard needle for administering vaccines by SC injection is a 25 or 26<br />

gauge needle, 16 mm in length.<br />

<strong>The</strong> immune response to vaccines inadvertently given IM rather than SC is<br />

unlikely to be affected. <strong>The</strong>refore it is usually not necessary to repeat doses in this<br />

instance.<br />

Intradermal injection technique<br />

For intradermal injection of BCG vaccine or Q fever skin test, a 26 or 27 gauge,<br />

10 mm needle is recommended. <strong>The</strong> intradermal injection technique requires<br />

special training, and should be performed only by a trained provider (see 4.20<br />

Tuberculosis and 4.15 Q fever).<br />

One brand of influenza vaccine (Intanza) is administered via the intradermal<br />

route. This vaccine is presented with a specifically designed needle and<br />

syringe called the Micro-Injection System and will deliver the 0.1 mL dose of<br />

vaccine into the dermal layer of the skin without the need for special training.<br />

Manufacturer’s instructions in the product packaging should be followed for<br />

correct administration.<br />

Table 2.2.2: Recommended needle size, length and angle for administering<br />

vaccines 10,16,18,21,25<br />

Age or size of child/adult Needle type Angle of needle<br />

insertion<br />

Infant, child or adult for IM vaccines<br />

Preterm babies (


Interruption to a vaccination<br />

If the process of administration of a vaccine given parenterally (IM or SC) is<br />

interrupted (e.g. by syringe–needle disconnection) and most of the dose has not<br />

been administered, the whole dose should be repeated as soon as practicable.<br />

If most of an oral rotavirus vaccine dose has been spat out or vomited within<br />

minutes of administration, a single repeat dose can be administered during<br />

the same visit. If an infant regurgitates or vomits only a small part of a dose<br />

of oral rotavirus vaccine, it is not necessary to repeat the dose. <strong>The</strong>refore, the<br />

regurgitated (and incomplete volume) dose is still considered as the valid dose<br />

(see 4.17 Rotavirus).<br />

2.2.6 Recommended injection sites<br />

<strong>The</strong> choice of injection sites depends primarily on the age of the person to be<br />

vaccinated. <strong>The</strong> two anatomical sites recommended as routine injection sites are<br />

the anterolateral thigh (Figures 2.2.5 and 2.2.6) and the deltoid muscle (Figure<br />

2.2.8). <strong>Immunisation</strong> service providers should ensure that they are familiar<br />

with the landmarks used to identify any anatomical sites used for vaccination.<br />

Photographs and diagrams are provided in this section, but are not a substitute<br />

for training. Further detail on identifying the recommended injection sites is<br />

provided in 2.2.8 Identifying the injection site.<br />

Infants


ut, if this site is used, the less locally reactogenic vaccines (e.g. MMR) should be<br />

given in the thigh.<br />

Children with congenital limb malformation or children in spica casts<br />

Children with congenital limb malformation(s) should receive their vaccines in<br />

an unaffected limb where possible. <strong>The</strong> ventrogluteal area can also be considered<br />

(see Figure 2.2.7 in 2.2.8 Identifying the injection site). 29<br />

Administration of vaccines to children in spica casts can be timed to occur when<br />

the cast is being changed. Parents should be informed of the importance of looking<br />

for any signs of swelling that may compromise circulation and, if this occurs,<br />

to seek advice from their physiotherapist or doctor as soon as possible. 29 Some<br />

resources suggest the use of the deltoid muscle as an alternative route for children<br />

in spica casts. If using this site, it is important to be aware of the radial nerve,<br />

which is located superficially near the deltoid in children


2.2.7 Positioning for vaccination<br />

It is important that infants and children do not move during injection of vaccines.<br />

However, excessive restraint can increase their fear and result in increased<br />

muscle tension. <strong>The</strong> following section describes a variety of positions that may be<br />

used for vaccinating different age groups.<br />

Infants


Positioning an infant on an examination table<br />

An alternative is to lay an infant on his/her back on an examination table, with<br />

the infant’s feet towards the immunisation service provider, and the parent/carer<br />

beside the provider to immobilise and distract the baby (see Figure 2.2.2).<br />

Keep the infant’s hip and knee flexed by cupping the patella in the non-injecting<br />

hand.<br />

<strong>The</strong> thumb and index finger of the non-injecting hand may be used to stabilise<br />

the hub of the needle once the needle has been inserted.<br />

Although the exact mechanism is unclear, recent studies have shown that placing<br />

a child in the supine position may result in more pain than if the child is held in<br />

an upright position. 15<br />

Figure 2.2.2: Positioning an infant on an examination table for vaccination<br />

Photo courtesy Dr Joanne Molloy, Victoria<br />

Prone position across the lap for ventrogluteal vaccination<br />

For ventrogluteal injection, position the child face-down across the parent/<br />

carer’s lap (see Figure 2.2.7 below). This allows the hips to be flexed and provides<br />

access to the ventrogluteal area.<br />

76 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Children ≥12 months of age<br />

Cuddle position for an older child<br />

Sit the child sideways on the lap of the parent/carer, with the arm to be injected<br />

held close to the child’s body while the other arm is tucked under the armpit and<br />

behind the back of the parent/carer.<br />

<strong>The</strong> child’s exposed arm should be secured at the elbow by the parent/carer, and<br />

the child’s legs should also be secured by the parent/carer (see Figure 2.2.3).<br />

Figure 2.2.3: Positioning an older child in the cuddle position<br />

Photo courtesy CHW Photography<br />

Straddle position<br />

An older child may be positioned facing the parent/carer with the legs straddled<br />

over the parent/carer’s lap. <strong>The</strong> child’s arms should be folded in front, with the<br />

parent/carer hugging the child’s body to the parent/carer’s chest. Alternatively<br />

the child may be positioned to ‘hug’ the parent/carer with the parent/carer’s<br />

arms holding the child’s arms in a reciprocal hug (see Figure 2.2.4). This position<br />

allows access to both deltoids and both anterolateral thighs.<br />

PART 2 VACCINATION PROCEDURES 77<br />

2.2 ADMINISTRATION<br />

OF VACCINES


Figure 2.2.4: Positioning a child in the straddle position<br />

Photo courtesy CHW photography<br />

Prone position across the lap for ventrogluteal vaccination<br />

For ventrogluteal injection, position the child face-down across the parent/<br />

carer’s lap (see Figure 2.2.7 below).<br />

Older children, adolescents and adults<br />

Solo sitting position for deltoid injections<br />

Most vaccines can be administered into the deltoid area. Adults should sit in<br />

a straight-backed chair, feet resting flat on the floor with forearms and hands<br />

in a relaxed position on the upper thighs. Keep the arms flexed at the elbow to<br />

encourage the deltoid muscle to relax.<br />

Encourage the shoulders to drop by asking the person to raise the shoulders<br />

up while taking a deep breath in and to drop them while breathing out fairly<br />

forcefully. Use distraction to keep muscles relaxed during the procedure, for<br />

example, have an interesting poster or similar for the person to concentrate on<br />

during the procedure and ask him/her to give you a detailed description of what<br />

can be seen.<br />

<strong>The</strong> ventrogluteal and vastus lateralis are alternative sites if needed (see 2.2.6<br />

Recommended injection sites and 2.2.8 Identifying the injection site).<br />

78 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


2.2.8 Identifying the injection site<br />

<strong>The</strong> choice of injection site depends on the age of the person to be vaccinated,<br />

and is discussed in 2.2.6 Recommended injection sites.<br />

<strong>The</strong> anterolateral thigh (vastus lateralis)<br />

• Make sure the infant’s nappy is undone to ensure the injection site is<br />

completely exposed and the anatomical markers can be easily identified by<br />

sight and palpation.<br />

• Position the leg so that the hip and knee are flexed and the vastus lateralis is<br />

relaxed (see Figure 2.2.6).<br />

• Identify the following anatomical markers: the upper marker is the midpoint<br />

between the anterior superior iliac spine and the pubic tubercle, and the<br />

lower marker is the upper part of the patella.<br />

• Draw an imaginary line between the two markers down the front of the<br />

thigh. <strong>The</strong> correct site for IM vaccination is lateral to the midpoint of this line,<br />

in the outer (anterolateral) aspect (see Figures 2.2.5 and 2.2.6).<br />

• Do not inject into the anterior aspect of the thigh where neurovascular<br />

structures can be damaged.<br />

Figure 2.2.5 Anatomical markers used to identify the vastus lateralis injection<br />

site (X) on the anterolateral thigh<br />

Level of greater<br />

trochanter<br />

Level of lateral<br />

femoral condyle<br />

Tensor fascia lata<br />

Injection site<br />

llio-tibial tract<br />

x<br />

Anterior superior<br />

iliac spine<br />

Pubic tubercle<br />

Femoral artery<br />

and vein<br />

Sartorius<br />

Rectus femoris<br />

Vastus lateralis<br />

Patella<br />

PART 2 VACCINATION PROCEDURES 79<br />

2.2 ADMINISTRATION<br />

OF VACCINES


Figure 2.2.6: <strong>The</strong> vastus lateralis injection site (X) on the anterolateral thigh<br />

Photo courtesy Lloyd Ellis, <strong>The</strong> Royal Children’s Hospital, Victoria<br />

<strong>The</strong> ventrogluteal area<br />

Note: This area should not be confused with the dorsogluteal area (buttock).<br />

<strong>The</strong> ventrogluteal area provides an alternative site for administering vaccines<br />

to a child of any age, especially when multiple injections at the same visit are<br />

required. <strong>The</strong> ventrogluteal area is relatively free of major nerves and blood<br />

vessels, and the area provides the greatest thickness of gluteal muscle. 35,36 <strong>The</strong>re is<br />

a relatively consistent thinness of subcutaneous tissue over the injection site. 36,37<br />

• Make sure the child’s nappy is undone to ensure the injection site is<br />

completely exposed and the anatomical markers can be easily identified by<br />

sight and palpation. Anatomical markers are the anterior superior iliac spine<br />

(ASIS), the greater trochanter of the femur and the iliac crest (see Figure<br />

2.2.7).<br />

• Place the child in a prone position (face-down) on the parent/carer’s lap<br />

or on the clinic table/bed, with the child’s arms tucked against their chest.<br />

Allow the child’s legs to dangle towards the floor (see Figure 2.2.7).<br />

• Ensure the knee and hip are turned inwards to encourage muscle relaxation<br />

at the injection site.<br />

• Use the injection site that is closest to you.<br />

80 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


• Place the palm over the greater trochanter (the uppermost bony prominence<br />

of the thigh bone), with the thumb pointing towards the umbilicus. Point<br />

the index finger towards the anterior superior iliac spine, and spread the<br />

middle finger so it aims at the iliac crest, thus creating a ‘V’ outlining the<br />

ventrogluteal triangular area. <strong>The</strong> injection site is at the centre of this area<br />

(see Figure 2.2.7).<br />

Figure 2.2.7: Anatomical markers used to identify the ventrogluteal injection<br />

site (X)<br />

ASIS = anterior superior iliac spine<br />

Photo courtesy of Dr Joanne Molloy, Victoria<br />

<strong>The</strong> deltoid area<br />

To locate the deltoid site for injection:<br />

• Expose the arm completely, from the top of the shoulder to the elbow; roll up<br />

the sleeve or remove the shirt if needed.<br />

• Locate the shoulder tip (acromion) and the muscle insertion at the middle of<br />

the humerus (deltoid tuberosity).<br />

• Draw an imaginary inverted triangle below the shoulder tip, using the<br />

identified anatomical markers (see Figure 2.2.8).<br />

<strong>The</strong> deltoid site for injection is halfway between the acromion and the deltoid<br />

tuberosity, in the middle of the muscle (triangle).<br />

PART 2 VACCINATION PROCEDURES 81<br />

2.2 ADMINISTRATION<br />

OF VACCINES


Figure 2.2.8: Anatomical markers used to identify the deltoid injection site<br />

Subcutaneous injection sites<br />

Subcutaneous injections should be administered either over the deltoid muscle<br />

or over the anterolateral thigh. <strong>The</strong>re are no studies that describe any specific<br />

differences in the technique used for an ‘SC injection’ compared with a ‘deep<br />

SC injection’. Figure 2.2.9 demonstrates the recommended technique for any SC<br />

injection.<br />

Figure 2.2.9: A subcutaneous injection into the deltoid area of the upper arm<br />

using a 25 gauge, 16 mm needle, inserted at a 45° angle<br />

Photo courtesy Jane Jelfs NCIRS<br />

82 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


2.2.9 Administering multiple vaccine injections at the same visit<br />

When sequentially administering multiple vaccines to children, give the most<br />

painful vaccine last (e.g. pneumococcal conjugate vaccine). Evidence suggests<br />

that this may decrease the overall pain response.<br />

<strong>The</strong> location of each separate injection given should be recorded, so that if a local<br />

adverse event occurs, the implicated vaccine(s) can be identified.<br />

Infants


• If the deltoid muscle mass is small:<br />

» give further injections into either anterolateral thigh (2.5 cm apart if two<br />

vaccines are given in the same thigh), or<br />

» give one injection into each ventrogluteal area.<br />

For younger children, the cuddle or straddle positions (Figures 2.2.3 and 2.2.4)<br />

are suitable for accessing multiple limbs during the one vaccination encounter.<br />

Simultaneous injections by two immunisation providers<br />

Currently there is insufficient evidence for or against having two immunisation<br />

providers administer vaccines at the same time rather than one vaccine after the<br />

other. 38,39 Two studies were unable to demonstrate a difference in pain response in<br />

the child between simultaneous administration and sequential administration. 38,39<br />

If multiple immunisation providers are available, the technique has been<br />

explained to the parent and the parent gives consent, then the vaccines may be<br />

administered simultaneously, providing different sites can be safely accessed.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

84 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


2.3 POST-VACCINATION<br />

2.3.1 Immediate after-care<br />

Immediately after vaccine administration:<br />

• dispose of clinical waste, including sharps and vaccine vials, at the point of<br />

use (refer to state/territory health authorities for management guidelines for<br />

the safe disposal of clinical waste or refer to the National Health and Medical<br />

Research Council’s <strong>Australian</strong> guidelines for the prevention and control of infection<br />

in healthcare) 1<br />

• cover the injection site quickly with a dry cotton ball and tape as needed<br />

• gently apply pressure for 1 or 2 minutes – do not rub the site as this will<br />

encourage the vaccine to leak back up the needle track, which can cause pain<br />

and may lead to local irritation<br />

• to distract the vaccinated person and reduce distress, immediately change the<br />

position of the child/person after completing the vaccination, for example,<br />

ask the parent/carer to put the infant over his/her shoulder and move<br />

around with the infant2 • remove the cotton wool after a few minutes and leave the injection site<br />

exposed to the air<br />

• record the relevant details of the vaccines given (see 2.3.3 Documentation of<br />

vaccination).<br />

<strong>The</strong> vaccinated person and/or parent/carer should be advised to remain in the<br />

vicinity for a minimum of 15 minutes after the vaccination. <strong>The</strong> area should be<br />

close enough to the immunisation service provider so that the vaccinated person<br />

can be observed and medical treatment provided rapidly if needed.<br />

Paracetamol is not routinely used before, or at the time of, vaccination, but may<br />

be recommended as required for fever or pain occurring following immunisation.<br />

Before departure, inform the vaccinated person or parent/carer, preferably in<br />

writing, of any expected adverse events following immunisation, and of the date<br />

of the next scheduled vaccination(s).<br />

Take the opportunity to check the vaccination status of other family members<br />

(as appropriate) and discuss any catch-up vaccination requirements and options<br />

available (this can also be done earlier in the visit).<br />

2.3.2 Adverse events following immunisation<br />

What are adverse events following immunisation?<br />

An adverse event following immunisation (AEFI) is any untoward medical<br />

occurrence that follows immunisation and does not necessarily have a causal<br />

relationship with the usage of the vaccine. <strong>The</strong> adverse event may be any<br />

unfavourable or unintended sign, abnormal laboratory finding, symptom or<br />

disease. 3 Such an event may be caused by the vaccine(s) or may occur by chance<br />

PART 2 VACCINATION PROCEDURES 85<br />

2.3 POST-VACCINATION


(i.e. it would have occurred regardless of vaccination). Most vaccine adverse<br />

events are minor, such as low-grade fever, and pain or redness at the injection<br />

site; these should be anticipated. 4 <strong>The</strong> frequency of adverse events has been<br />

classified by regulatory agencies, and is often reported in clinical trials as follows:<br />

very common (>10% of persons vaccinated), common (1–10%), uncommon (0.1–<br />


Management of an immediate adverse event following immunisation<br />

<strong>The</strong> vaccinated person should remain under observation for a short interval<br />

to ensure that they do not experience an immediate adverse event. It is<br />

recommended that vaccinated persons remain in the vicinity of the place of<br />

vaccination for at least 15 minutes. Severe anaphylactic reactions usually have<br />

a rapid onset; life-threatening adverse events are most likely to begin within 15<br />

minutes of vaccination.<br />

<strong>The</strong> most serious immediate AEFI is anaphylaxis. However, in adults and older<br />

children, the most common immediate adverse event is a vasovagal episode<br />

(fainting), either immediately or soon after vaccination. Because fainting<br />

after vaccination can lead to serious consequences, anyone who complains of<br />

giddiness or light-headedness before or after vaccination should be advised to lie<br />

down until free of symptoms.<br />

Anaphylaxis and vasovagal episodes<br />

Anaphylaxis following routine vaccination is very rare, but can be fatal. 6 All<br />

immunisation service providers must be able to recognise all the symptoms<br />

and signs of anaphylaxis and distinguish between anaphylaxis, convulsions<br />

and fainting. <strong>The</strong> features listed in Table 2.3.1 may be useful in differentiating<br />

between fainting (vasovagal episode) and anaphylaxis.<br />

Anaphylaxis is a severe adverse event of rapid onset, characterised by<br />

sudden respiratory compromise and/or circulatory collapse. 7 Early signs<br />

include involvement of the skin (e.g. generalised erythema, urticaria and/or<br />

angioedema) and/or gastrointestinal tract (e.g. diarrhoea, vomiting). In severe<br />

cases, there is circulatory collapse with alteration in the level of consciousness,<br />

hypotension and weak or absent pulses, and/or marked respiratory compromise<br />

from upper airway oedema or bronchospasm.<br />

Fainting (vasovagal episode) is relatively common after vaccination of adults and<br />

adolescents, but infants and children rarely faint. Sudden loss of consciousness<br />

in young children should be presumed to be anaphylaxis, particularly if a strong<br />

central pulse is absent. A strong central pulse (e.g. carotid) persists during a faint<br />

or convulsion.<br />

If a diagnosis of anaphylaxis is suspected, treatment, including administration<br />

of adrenaline, should be instituted promptly8 (see ‘Management of anaphylaxis’<br />

below). Under-treatment of anaphylaxis is more harmful, and potentially lifethreatening,<br />

than over-treatment of a mild or moderate allergic reaction. 9<br />

PART 2 VACCINATION PROCEDURES 87<br />

2.3 POST-VACCINATION


Table 2.3.1: Clinical features that may assist differentiation between a<br />

vasovagal episode and anaphylaxis<br />

Vasovagal<br />

episode<br />

Onset Immediate,<br />

usually within<br />

minutes of, or<br />

during, vaccine<br />

administration<br />

Symptoms/<br />

Signs<br />

Respiratory Normal<br />

respiration; may<br />

be shallow, but<br />

not laboured<br />

Cardiovascular Bradycardia,<br />

weak/absent<br />

peripheral pulse,<br />

strong carotid<br />

pulse<br />

Hypotension –<br />

usually transient<br />

and corrects in<br />

supine position<br />

Loss of<br />

consciousness<br />

– improves<br />

once supine or<br />

in head-down<br />

position<br />

Skin Generalised<br />

pallor, cool,<br />

clammy skin<br />

Gastrointestinal Nausea/<br />

vomiting<br />

Neurological † Feels faint, lightheaded<br />

88 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Anaphylaxis*<br />

Usually within 15 minutes,<br />

but can occur within hours, of<br />

vaccine administration<br />

Cough, wheeze, hoarseness,<br />

stridor, or signs of respiratory<br />

distress (e.g. tachypnoea,<br />

cyanosis, rib recession)<br />

Upper airway swelling (lip,<br />

tongue, throat, uvula or larynx)<br />

Tachycardia, weak/absent<br />

carotid pulse<br />

Hypotension – sustained and<br />

no improvement without<br />

specific treatment (Note: in<br />

infants and young children,<br />

limpness and pallor are signs<br />

of hypotension)<br />

Loss of consciousness – no<br />

improvement once supine or in<br />

head-down position<br />

Pruritus (skin itchiness),<br />

generalised skin erythema<br />

(redness), urticaria (weals)<br />

or angioedema (localised<br />

or general swelling of the<br />

deeper layers of the skin or<br />

subcutaneous tissues)<br />

Abdominal cramps, diarrhoea,<br />

nausea and/or vomiting<br />

Sense of severe anxiety and<br />

distress<br />

* Modified from <strong>The</strong> Brighton Collaboration Case Definition Criteria for Anaphylaxis. 6<br />

† Neurological symptoms are not listed in the Brighton case definition criteria for anaphylaxis; 6<br />

however, symptoms of anxiety and distress, including feelings of impending doom, are reported<br />

in persons experiencing anaphylaxis. 7,10


Management of anaphylaxis<br />

Rapid IM administration of adrenaline is the cornerstone of treatment of<br />

anaphylaxis. Adrenaline is life saving and must be used promptly. 8<br />

Anaphylaxis occurs without warning, usually within 15 minutes of giving a<br />

vaccine. A protocol for the management of anaphylaxis, adrenaline and 1 mL<br />

syringes must always be immediately at hand whenever vaccines are given.<br />

• If the patient is unconscious, lie him/her on the left side and position to keep<br />

the airway clear.<br />

• If the patient is conscious, lie him/her supine in ‘head-down and feet-up’<br />

position (unless this results in breathing difficulties).<br />

• If there are any respiratory and/or cardiovascular symptoms or signs of<br />

anaphylaxis, give adrenaline by IM injection into the anterolateral thigh<br />

(see ‘Use of adrenaline’ below for dosage). Adrenaline is not required for<br />

generalised non-anaphylactic reactions (such as skin rash or angioedema).<br />

If in doubt, IM adrenaline should be given. No serious or permanent harm<br />

is likely to occur from mistakenly administering adrenaline to an individual<br />

who is not experiencing anaphylaxis. 11<br />

• Call for assistance. Never leave the patient alone.<br />

• If oxygen is available, administer by facemask at a high flow rate.<br />

• If there is no improvement in the patient’s condition within 5 minutes, repeat<br />

doses of adrenaline every 5 minutes until improvement occurs.<br />

• Check breathing; if absent, commence basic life support or appropriate<br />

cardiopulmonary resuscitation (CPR), as per the <strong>Australian</strong> Resuscitation<br />

Council guideline12 (available at www.resus.org.au/policy/guidelines).<br />

• In all cases, transfer the person to hospital for further observation and<br />

treatment.<br />

• Complete full documentation of the event, including the time and dose(s) of<br />

adrenaline given.<br />

Antihistamines and/or hydrocortisone are not recommended for the emergency<br />

management of anaphylaxis.<br />

Use of adrenaline<br />

<strong>The</strong> use of 1:1000 adrenaline is recommended because it is universally available.<br />

Adrenaline 1:1000 (one in one thousand) contains 1 mg of adrenaline per mL of<br />

solution in a 1 mL glass vial. Adrenaline 1 in 10 000 is no longer recommended<br />

for the treatment of anaphylaxis. A 1 mL syringe should be used to improve the<br />

accuracy of measurement when drawing up small doses of adrenaline.<br />

<strong>The</strong> recommended dose of 1:1000 adrenaline is 0.01 mL/kg body weight<br />

(equivalent to 0.01 mg/kg or 10 µg/kg) up to a maximum of 0.5 mL, given by<br />

deep IM injection preferably in the anterolateral (upper outer) thigh. <strong>The</strong> anterolateral<br />

thigh is the preferred site because there is a more predictable dispersal of<br />

PART 2 VACCINATION PROCEDURES 89<br />

2.3 POST-VACCINATION


adrenaline from this site. 13 Administration of adrenaline in the anterolateral<br />

thigh is also in accordance with recommendations from various emergency<br />

medicine, anaesthetic and immunology professional bodies. 9<br />

Adrenaline 1:1000 must not be administered intravenously.<br />

Table 2.3.2 lists the dose of 1:1000 adrenaline to be used if the exact weight of<br />

the person is not known.<br />

<strong>The</strong> dose of 1:1000 (one in one thousand) adrenaline may be repeated every<br />

5 minutes, as necessary, until there is clinical improvement.<br />

Table 2.3.2: Doses of intramuscular 1:1000 (one in one thousand) adrenaline for<br />

anaphylaxis* 14<br />

Approximate age and weight Adrenaline dose<br />

12 years and adult (over 50 kg) 0.5 mL<br />

* Modified from insert published in <strong>Australian</strong> Prescriber 14 (available at www.australianprescriber.<br />

com/magazine/34/4/article/1210.pdf). Endorsed by the Australasian Society of Clinical<br />

Immunology and Allergy, the Royal Australasian College of Physicians, the Royal <strong>Australian</strong><br />

College of General Practitioners, the Australasian College for Emergency Medicine, the Royal<br />

<strong>Australian</strong> and New Zealand College of Radiologists, the Internal Medicine Society of Australia<br />

and New Zealand, and the <strong>Australian</strong> Dental Association.<br />

Use of adrenaline autoinjectors for anaphylaxis treatment<br />

Adrenaline autoinjectors, EpiPen or Anapen, are devices that administer a single,<br />

pre-measured dose of adrenaline. <strong>The</strong>y are designed for use by any person,<br />

whether medically trained or not. Clear instructions on correct use are provided<br />

on the barrel and in the packaging of these devices. <strong>The</strong>y are designed to be<br />

administered in the mid-outer thigh.<br />

Autoinjectors are usually recommended or prescribed for an individual who is<br />

at risk of anaphylaxis due to an existing allergy or where skin testing indicates<br />

a high risk of an allergic reaction on exposure to an allergen. If a patient who<br />

carries an autoinjector device develops anaphylaxis post vaccination, it is<br />

appropriate to use their autoinjector to administer adrenaline.<br />

90 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Autoinjectors are generally not appropriate for inclusion in first aid kits for<br />

general use, due to several limitations:<br />

• they are single-use only<br />

• they are dose-specific<br />

» EpiPen Jr or Anapen Jr containing 150 µg of adrenaline are recommended<br />

for children weighing between 10 kg and 20 kg<br />

» EpiPen or Anapen containing 300 µg of adrenaline are recommended for<br />

children and adults weighing over 20 kg<br />

• multiple pens would be required to allow for repeat dosing and varying<br />

ages/weights of patients, and shelf-life is limited to 1 to 2 years maximum.<br />

Autoinjectors are not recommended for use in children weighing less than 10 kg.<br />

Common adverse events following immunisation and their management<br />

Commonly occurring AEFI are described in the table Comparison of the effects of<br />

diseases and the side effects of NIP vaccines inside the back cover of this <strong>Handbook</strong><br />

and in the disease-specific chapters in Part 4.<br />

<strong>The</strong> most commonly encountered adverse events are local reactions related to<br />

vaccine injection(s), such as pain, redness, itching, swelling or burning at the<br />

injection site. <strong>The</strong>se are to be expected, are generally mild and usually last for<br />

1 to 2 days. Injection site nodules are also relatively common. <strong>The</strong>y are fibrous<br />

remnants of the body’s interaction with the vaccine components (usually an<br />

adjuvant) in the muscle. <strong>The</strong>y may remain for many weeks after vaccination and<br />

do not require any specific treatment.<br />

Low-grade fever and tiredness (malaise), lasting a few days, are also common<br />

after many vaccines. <strong>The</strong>se responses are usually mild and self-limiting, and<br />

generally do not require specific treatment.<br />

Routine use of paracetamol at the time of, or immediately after, vaccination is<br />

not recommended. However, if an infant, child or adult has a fever of >38.5°C<br />

following vaccination or has pain at the injection site, paracetamol can be given.<br />

<strong>The</strong> dose of paracetamol for an infant or child up to 12 years of age is<br />

15 mg/kg/dose, up to a maximum dose of 60 mg/kg per day in four divided<br />

doses. Adults and children aged ≥12 years can receive 500 to 1000 mg every 4 to 6<br />

hours; dosage must not exceed 4 g in 24 hours. Paracetamol should not be given<br />

for more than 48 hours without seeking medical advice. 15<br />

If patients exhibit unexpected, serious or prolonged adverse symptoms or signs<br />

following immunisation, medical advice should be sought. <strong>The</strong> symptoms and<br />

signs from medical illness unrelated to vaccination can sometimes be attributed<br />

to a recent immunisation and should be investigated and managed accordingly.<br />

PART 2 VACCINATION PROCEDURES 91<br />

2.3 POST-VACCINATION


Uncommon/rare adverse events following immunisation<br />

Some vaccines have been shown to cause uncommon or rare serious adverse<br />

events, although the rate of vaccine adverse events is usually hundreds to<br />

thousands times less frequent than the disease complications. Information on the<br />

benefits compared with risks of immunisation is always taken into account when<br />

making recommendations for vaccine use. It is important to provide persons<br />

to be vaccinated or their parent/carer with advice regarding known, but rare,<br />

adverse events following immunisation, and to place the advice in the context of<br />

the benefits of vaccination (see 3.3.1 Vaccination of persons who have had an adverse<br />

event following immunisation).<br />

If a patient has experienced a serious or uncommon/rare AEFI, it is important<br />

that they or their immunisation service provider seek advice from a specialist<br />

immunisation clinic or contact state/territory health authorities for more<br />

information regarding the need for further investigation and management (see<br />

Appendix 1 Contact details for <strong>Australian</strong>, state and territory government health<br />

authorities and communicable disease control). This will enable an assessment to<br />

determine the relationship to vaccination, consideration of the benefits and risks<br />

of further vaccination, and planning for receiving additional doses of that or<br />

other vaccines, as appropriate. Persons who have had a serious adverse event<br />

following immunisation (other than a contraindication, such as the confirmed<br />

identity of the vaccine component that triggered anaphylaxis) can usually<br />

subsequently be vaccinated under close medical supervision. For more detailed<br />

information see 3.3.1 Vaccination of persons who have had an adverse event following<br />

immunisation.<br />

Examples of uncommon and rare adverse events are given below. It is important<br />

to remember that, although these events are uncommon or rare, they are still<br />

not necessarily causally related to vaccination, even if they occur following<br />

vaccination.<br />

• Febrile convulsions are a relatively common response to fever of any cause<br />

in young children, particularly in those aged


Australia. 16 This vaccine is no longer registered for use in this age group. An<br />

excess risk of fever and febrile convulsions was not observed with the other<br />

influenza vaccines given to children. 16,17<br />

• Brachial neuritis (inflammation of a nerve in the arm, causing weakness<br />

or numbness) has been described following the administration of tetanus<br />

toxoid-containing vaccines, with an estimated excess risk of approximately<br />

0.5–1 in 100 000 doses in adults. 5,18 Case reports of brachial neuritis following<br />

administration of other vaccines, including HPV vaccines, 19 are rare and a<br />

causal relationship has not been established. 20<br />

• Oral rotavirus vaccines are associated with a small increased risk of<br />

intussusception (IS), a rare form of bowel blockage caused by telescoping<br />

of the intestine into itself. This risk appears to be particularly in the 7 days<br />

following the 1st vaccine dose; however, a smaller increased risk in the week<br />

following the 2nd dose has also been reported. 21-23 It is not currently clear<br />

whether there is an overall increase in the risk of IS above that which would<br />

be expected in the 1st year of infancy without vaccine use. <strong>The</strong> increased risk<br />

represents approximately 6 additional cases of intussusception among every<br />

100 000 infants vaccinated, or 18 additional cases per year in Australia. 23<br />

Children who have had IS are recommended to not receive rotavirus vaccine<br />

(see 4.17 Rotavirus).<br />

• Anaphylaxis following receipt of vaccines has been reported, but generally<br />

occurs very rarely. 24 For example, the estimated incidence rate of anaphylaxis<br />

following 4vHPV vaccine in Australia as at June 2010 was 2.6 anaphylaxis<br />

episodes per million doses of vaccine distributed. 25 This is within the<br />

same rate range as for other vaccines given to children and adolescents in<br />

international studies, such as hepatitis B vaccine, which is associated with<br />

anaphylaxis in approximately 1 in 1.1 million doses distributed. 26 (For more<br />

information on the management of immediate AEFI/anaphylaxis, see above.)<br />

• Hypotonic-hyporesponsive episode (HHE) is the sudden onset of pallor<br />

or cyanosis, limpness (muscle hypotonia), and reduced responsiveness<br />

or unresponsiveness occurring after vaccination, where no other cause is<br />

evident such as a vasovagal episode or anaphylaxis. <strong>The</strong> episode usually<br />

occurs 1 to 48 hours after vaccination and resolves spontaneously. <strong>The</strong>re are<br />

no known long-term side effects from HHE. 27,28 In Australia during 2009,<br />

3.2 cases of HHE were reported per 100 000 doses of DTPa-containing vaccine<br />

given to children


Events where evidence demonstrates no causal link with immunisation<br />

Since vaccines are mainly given to healthy people, a range of conditions that<br />

occur after a vaccine dose may be attributed to vaccination. This is particularly so<br />

for illnesses that are complex and have an unknown or unclear cause. As many<br />

of these illnesses are rare and/or manifest months to years after vaccination,<br />

they are difficult to study in randomised controlled clinical trials, which are<br />

typically conducted before vaccines are registered for use. However, there is<br />

strong epidemiological evidence, usually derived from multiple well-conducted<br />

post-marketing studies, that indicates there is no causal association between<br />

immunisation and many diseases/conditions in which vaccines were suggested<br />

to have been involved.<br />

Examples of events unrelated to vaccination include:<br />

• sudden infant death syndrome (SIDS) and any vaccine31-33 • autism and MMR vaccine34-39 • multiple sclerosis and hepatitis B vaccine40-43 • inflammatory bowel disease and MMR vaccine44 • diabetes and Hib vaccine45-47 • asthma and any vaccine. 48<br />

Despite this evidence, patients/parents seeking further advice should discuss<br />

this with their immunisation provider or could be referred to a specialist<br />

immunisation clinic for further reassurance (see Appendix 1 Contact details<br />

for <strong>Australian</strong>, state and territory government health authorities and communicable<br />

disease control).<br />

Reporting adverse events following immunisation<br />

Surveillance for adverse events following immunisation is an integral part<br />

of the <strong>Australian</strong> National <strong>Immunisation</strong> Program, and underpins the safe<br />

use of all vaccines in Australia. Surveillance of AEFI aims to detect changes<br />

in the rates of known adverse events, any unrecognised or unexpected<br />

adverse events, or adverse events that result from program errors, such as<br />

incorrect vaccine schedule, delivery or storage. It is very important that<br />

all immunisation service providers report AEFI, particularly if serious or<br />

unexpected, as this will enable vaccine safety issues to be identified and<br />

managed appropriately as soon as possible. For example, reporting of AEFI by<br />

immunisation service providers in Australia in 2010 resulted in the detection of<br />

an unexpectedly high rate of fever and febrile convulsions in young children,<br />

associated with the use of one brand of seasonal influenza vaccine. 49,50 All<br />

reported AEFI are included in the Adverse Drug Reactions System (ADRS)<br />

database of the <strong>The</strong>rapeutic Goods Administration (TGA). For details on how<br />

to report AEFI, see the next section below.<br />

94 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Any serious or unexpected adverse event following immunisation should<br />

be promptly reported. Providers should use clinical judgment in deciding<br />

which adverse events to report and parents/carers should be encouraged<br />

to notify the immunisation service provider or health authorities of any<br />

untoward medical occurrence that follows immunisastion.<br />

No time limit has been set to report AEFI; however, timely notification of adverse<br />

events, particularly rapid reporting of serious events, is important to identify any<br />

potential concerns. Notification does not necessarily imply a causal association<br />

with vaccination, as some events may occur coincidentally following vaccination.<br />

Any event that is suspected of being related to vaccination can be reported.<br />

All identifying information relating to the reporter and patient is kept strictly<br />

confidential. Any person, medical or non-medical, including providers who did<br />

not give the vaccine(s), can report an AEFI; however, it is very important that as<br />

much detail as possible is provided on all reports.<br />

In addition to reporting of AEFI, immunisation service providers may need<br />

to provide additional clinical management and advice regarding future<br />

vaccination(s) for their patient and may require expert advice. Information about<br />

specialist immunisation clinics, or the contact details for paediatricians or medical<br />

specialists with experience in management of patients with AEFI, are usually<br />

available from state and territory health authorities (see Appendix 1 Contact details<br />

for <strong>Australian</strong>, state and territory government health authorities and communicable<br />

disease control) and from the Immunise Australia website (www.immunise.health.<br />

gov.au). For more information on managing common and rare AEFI,<br />

see above and also 3.3.1 Vaccination of persons who have had an adverse event<br />

following immunisation.<br />

How to report adverse events following immunisation<br />

AEFI are notifiable via different routes; immunisation service providers should<br />

be aware of the method of reporting for their location. In most jurisdictions<br />

(the <strong>Australian</strong> Capital Territory, New South Wales, the Northern Territory,<br />

Queensland, South Australia, Victoria and Western Australia), AEFI should be<br />

reported directly to the relevant state/territory health authority (see Table 2.3.3).<br />

AEFI notified to these state and territory health departments are then forwarded<br />

to the TGA, who manage the ADRS database, which includes all adverse reaction<br />

reports related to drugs and vaccines. Reporting can also be done directly to the<br />

TGA as described below.<br />

PART 2 VACCINATION PROCEDURES 95<br />

2.3 POST-VACCINATION


Table 2.3.3: Contact information for notification of adverse events<br />

following immunisation<br />

State/Territory Report adverse events to Contact information<br />

<strong>Australian</strong> Capital<br />

Territory<br />

ACT Health Department 02 6205 2300<br />

New South Wales NSW Public Health Units 1300 066 055<br />

(for connection to Public<br />

Health Unit)<br />

Northern Territory NT Department of Health 08 8922 8044<br />

Queensland Queensland Health Complete an AEFI initial report<br />

form, available at: www.health.<br />

qld.gov.au/immunisation or by<br />

phoning 07 3328 9888.<br />

Fax the completed form to the<br />

number provided on the form.<br />

South Australia SA Health 1300 232 272<br />

www.sahealth.sa.gov.au<br />

Tasmania Direct to the TGA 1800 044 114<br />

or complete the ‘Blue card’<br />

reporting form (see below)<br />

Victoria SAEFVIC 03 9345 4143 or online at www.<br />

saefvic.org.au<br />

Western Australia State Health Department,<br />

WAVSS<br />

96 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

08 9321 1312 or online at wavss.<br />

health.wa.gov.au<br />

Alternatively, reporting directly to the TGA can be done by any person in<br />

any jurisdiction. Reports are submitted using the ‘Blue card’ adverse reaction<br />

reporting form. Paper copies of the ‘Blue card’ are available from:<br />

Office of Product Review<br />

<strong>The</strong>rapeutic Goods Administration<br />

Reply Paid 100<br />

Woden ACT 2606<br />

Telephone: 1800 044 114<br />

or online at www.tga.gov.au/safety/problem-medicines-forms-bluecard.htm.<br />

Alternatively, the adverse reaction reporting form can be completed and<br />

submitted online via the TGA website at www.ebs.tga.gov.au/ebs/ADRS/<br />

ADRSRepo.nsf?OpenDatabase.


Consumers and immunisation service providers can also report AEFI (and<br />

adverse drug reactions) via the national Adverse Medicines Events telephone<br />

reporting line on 1300 134 237. This service is operated by the National<br />

Prescribing Service (NPS) (www.nps.org.au) and funded by the <strong>Australian</strong><br />

Government through the Department of Health and Ageing.<br />

<strong>The</strong> TGA, in turn, forwards copies of individual reports of AEFI from vaccines<br />

on the NIP schedule back to state/territory health departments for their<br />

information.<br />

Information on AEFI reports to the TGA from all sources are aggregated, and<br />

detailed information on AEFI reporting rates and trends in AEFI are published on<br />

a 6-monthly basis in the journal Communicable Diseases Intelligence (www.health.<br />

gov.au/internet/main/publishing.nsf/content/cda-pubs-cdi-cdiintro.htm). 24,51<br />

2.3.3 Documentation of vaccination<br />

It is essential that immunisation service providers ensure there is appropriate<br />

documentation of all vaccinations given to persons of any age. <strong>The</strong>re are a<br />

number of ways in which this should be done.<br />

All vaccines administered to children should be documented in the child’s<br />

clinical file and the individual child health record that is established for all<br />

newborn infants. This record should be kept by the parent/carer and presented<br />

every time the child is seen by a health professional.<br />

Vaccines administered to adolescents and adults should be recorded in both the<br />

vaccinated person’s clinical file and the personal health record, or individual<br />

record, of vaccination. <strong>The</strong> following details should be recorded:<br />

• the person’s full name and date of birth<br />

• the details of the vaccine given, including the brand name, batch<br />

number and dose number<br />

• the date and time of vaccination<br />

• the site of administration<br />

• the name of the person providing the vaccination<br />

• the date the next vaccination is due.<br />

Some state/territory health departments also have specific requirements for<br />

documentation of vaccines administered to healthcare workers/healthcare<br />

students undertaking work or clinical placement within state/territory health<br />

facilities. Refer to the relevant state/territory health department for further<br />

details (see Appendix 1).<br />

<strong>Immunisation</strong> service providers should also report vaccination details to the<br />

appropriate immunisation register(s) – discussed in detail below.<br />

PART 2 VACCINATION PROCEDURES 97<br />

2.3 POST-VACCINATION


2.3.4 <strong>Immunisation</strong> registers<br />

<strong>Australian</strong> Childhood <strong>Immunisation</strong> Register<br />

<strong>The</strong> <strong>Australian</strong> Childhood <strong>Immunisation</strong> Register (ACIR) is a national database<br />

for recording details of vaccinations given to children


When relevant, immunisation service providers should complete either the<br />

‘Medical Contraindication’ or other objection forms and forward these to<br />

the ACIR.<br />

For further information about the ACIR and reporting vaccination information,<br />

see ‘<strong>The</strong> ACIR on the Internet’ below. In addition, assistance on any reporting<br />

issues can be obtained from the ACIR Enquiry Line, 1800 653 809 (free call).<br />

<strong>The</strong> <strong>Australian</strong> Childhood <strong>Immunisation</strong> Register on the Internet<br />

<strong>The</strong> Department of Human Services (DHS) website (www.humanservices.<br />

gov.au/customer/services/medicare/medicare-online-services) houses ACIR<br />

information and resources. <strong>The</strong> website has a general information area for<br />

individuals and families, a general information area for health professionals and<br />

a secure area for approved immunisation service providers only.<br />

<strong>The</strong> secure ACIR Internet site (https://www1.medicareaustralia.gov.au/ssl/<br />

acircirssamn) allows approved immunisation service providers to obtain a range<br />

of statistical and identified reports. Depending on the access level granted to the<br />

provider, these reports enable approved providers to view a child’s vaccination<br />

details, record vaccination information and access a range of other reports.<br />

All health professionals with access to Health Professional Online Services<br />

(HPOS) can access the ACIR Internet site through this platform. In addition,<br />

immunisation service providers can register for access to the secure ACIR<br />

Internet site only by completing the online request form, available from the<br />

Department of Human Services website. Further information or assistance may<br />

be obtained by calling the ACIR Internet Helpline on 1300 650 039.<br />

<strong>Immunisation</strong> History Statement<br />

<strong>Immunisation</strong> History Statements contain details of all vaccines administered<br />

to a child that are recorded on the ACIR and list the vaccines that are next due.<br />

<strong>The</strong>se Statements are automatically generated when a child turns 18 months of<br />

age, 5 years of age, and on completion of the childhood vaccination schedule.<br />

<strong>The</strong>se Statements will be mailed to the address most recently recorded by<br />

Medicare for that child.<br />

Parents/carers can also get an <strong>Immunisation</strong> History Statement at any other<br />

time:<br />

• online at www.humanservices.gov.au/customer/services/medicare/<br />

australian-childhood-immunisation-register<br />

• from their local DHS Service Centres<br />

• by calling 1800 653 809 (free call).<br />

PART 2 VACCINATION PROCEDURES 99<br />

2.3 POST-VACCINATION


<strong>Immunisation</strong> History Statements can be used to assist in recalling vaccination<br />

history when required. For example:<br />

• for school enrolment – a sentence will be displayed at the bottom of the<br />

Statement that says the child has received all the vaccinations required by<br />

5 years of age<br />

• to determine eligibility for the Child Care Benefit and the Family Tax<br />

Benefit Part A supplement – this requires that children are assessed as<br />

fully immunised; this replaced the Maternity <strong>Immunisation</strong> Allowance on<br />

1 July 2012.<br />

<strong>The</strong> ACIR Enquiry Line can be contacted on 1800 653 809 (free call) and any<br />

record held for a person who is now ≥7 years of age can be made available to an<br />

immunisation service provider or parent/carer.<br />

Recording details of a deceased child<br />

<strong>The</strong> ACIR should be notified of the death of a child to prevent an <strong>Immunisation</strong><br />

History Statement being sent to bereaved parents/carers. Advice of a child’s<br />

death can be provided to the ACIR by calling 1800 653 809 (free call), or by<br />

sending details on practice stationery. Details should include the child’s name,<br />

address, date of birth, Medicare number and date of death.<br />

Children who have moved to live overseas<br />

A child who has moved overseas can be removed from the ACIR by sending<br />

details to the ACIR by fax, phone or secure site email. This prevents the child’s<br />

name continuing to appear on ACIR reports of overdue children.<br />

Children born overseas who have moved to live in Australia permanently<br />

A child born overseas who has moved permanently to Australia will be<br />

automatically added to the ACIR upon enrolment with Medicare. Children<br />

residing temporarily in Australia are not included on the ACIR.<br />

Ascertaining individual vaccination status<br />

Parents/carers can telephone the ACIR on 1800 653 809 (free call) for information<br />

about their child’s vaccination status, regardless of where the child’s vaccination<br />

was given. <strong>Immunisation</strong> service providers can also request a child’s vaccination<br />

status by telephone.<br />

Vaccination coverage and other reports<br />

ACIR reports assess progress towards national targets, help to identify areas with<br />

low vaccination levels, and assist in planning vaccination programs.<br />

Practices can receive quarterly reports on vaccination coverage for children<br />

within their practice. Other reports, including those that identify a child’s<br />

vaccinations and due/overdue details, are available through the secure area<br />

of the ACIR Internet site (https://www1.medicareaustralia.gov.au/ssl/<br />

acircirssamn) to approved immunisation service providers.<br />

100 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


National Human Papillomavirus Vaccination Program Register<br />

<strong>The</strong> National HPV Vaccination Program Register (NHVPR), also referred to<br />

as the ‘HPV Register’, was established following the passing of the National<br />

Health Amendment Act (National HPV Vaccination Program Register) Bill in<br />

2007. Establishment and maintenance of the HPV Register facilitates the<br />

implementation of the National HPV Vaccination Program funded by the<br />

<strong>Australian</strong> Government. <strong>The</strong> HPV Register is run by the Victorian Cytology<br />

Service, together with the Department of Health and Ageing. It plays an essential<br />

role in monitoring and evaluating the HPV Vaccination Program by recording<br />

information about HPV vaccine doses administered in Australia.<br />

Reporting to the HPV Register<br />

Details on HPV vaccinations given in the community are provided to the HPV<br />

Register by the immunisation service provider who administers the vaccine.<br />

Vaccination details may be submitted electronically, via data uploads or direct<br />

entry using the secure website, or in hard copy, using one of the approved<br />

notification forms. <strong>Immunisation</strong> service providers in Queensland and the<br />

Northern Territory report data to the HPV Register via their state/territory<br />

health authority. <strong>Immunisation</strong> service providers wishing to submit vaccination<br />

data electronically need to be approved and registered with the HPV Register in<br />

order to notify administered doses. General practitioners are required to register<br />

with the HPV Register in order to notify administered doses. Further information<br />

about registration and notification procedures is available from the HPV Register<br />

website (www.hpvregister.org.au) or by phoning 1800 478 734 (1800 HPV REG).<br />

HPV vaccination coverage and other reports<br />

For immunisation service providers, the HPV Register has developed overdue<br />

HPV vaccine dose reports for their patients, which are available online via the<br />

secure website. De-identified HPV vaccination coverage data and other reports<br />

have also been developed to inform policy making, and support program<br />

delivery and approved research. National coverage data are made publicly<br />

available via the Immunise Australia website (www.immunise.health.gov.au).<br />

HPV Register statements<br />

<strong>The</strong> HPV Register sends Completion Statements and History/Reminder<br />

Statements. <strong>Immunisation</strong> History Statements, containing details of the<br />

vaccinations recorded on the HPV Register, are sent to persons who are overdue<br />

for HPV vaccination within the school-based program. Completion Statements<br />

are sent to persons who have completed the 3-dose HPV vaccination course.<br />

Vaccinated persons and parents/guardians can request a statement at any time<br />

by phoning 1800 478 734 (1800 HPV REG). In the event that booster doses are<br />

required in future, all eligible persons will be notified by the HPV Register.<br />

PART 2 VACCINATION PROCEDURES 101<br />

2.3 POST-VACCINATION


HPV vaccination status<br />

Vaccinated persons and parents/guardians can phone the HPV Register on<br />

1800 478 734 (1800 HPV REG) to obtain their or their child’s HPV vaccination<br />

status. <strong>Immunisation</strong> service providers can also request a person’s vaccination<br />

status by phone or can view these online if they are registered with the HPV<br />

Register. <strong>The</strong> HPV Register initially only recorded vaccinations for females, but<br />

from <strong>2013</strong> will also record vaccinations given to males.<br />

HPV Register secure website<br />

<strong>The</strong> HPV Register secure website allows registered and approved immunisation<br />

service providers access to the live national HPV Register database to view a<br />

patient’s vaccination history as well as access to overdue dose reports. Further<br />

information on how to request access to the HPV Register secure website can be<br />

found on the health professionals page of the HPV Register website<br />

(www.hpvregister.org.au) or by phoning 1800 478 734 (1800 HPV REG).<br />

Other immunisation registers<br />

<strong>The</strong> <strong>Australian</strong> Q Fever Register (www.qfever.org), established by Meat and<br />

Livestock Australia (MLA), has records of receipt of Q fever vaccination for some<br />

individuals, which can be accessed by registered users (see also 4.15 Q fever).<br />

State/territory government health departments maintain records of<br />

immunisations provided through school-based vaccination programs.<br />

Information on how to access records of vaccines received through school-based<br />

vaccination programs can be obtained from state/territory government health<br />

departments (see Appendix 1 Contact details for <strong>Australian</strong>, state and territory<br />

government health authorities and communicable disease control).<br />

Some state/territory governments operate a jurisdictional immunisation register,<br />

though the scope of these varies.<br />

Queensland<br />

<strong>The</strong> Vaccine Information and Vaccine Administration System (VIVAS) is a<br />

database of vaccination events for all children up to 10 years of age, adolescents<br />

and (some) adults in Queensland who are vaccinated with nationally- or statefunded<br />

vaccines.<br />

<strong>Immunisation</strong> service providers in Queensland are encouraged to report all<br />

vaccinations, either directly to VIVAS via the Queensland Health Vaccination<br />

Record form or using practice software to electronically transfer data (via the<br />

ACIR) to Queensland Health. Vaccination Record forms can be posted reply<br />

paid to VIVAS or faxed directly. Providers reporting to VIVAS should do so at<br />

least once a week to ensure the supply of data is not delayed and is available<br />

for the purposes of calculating parental and provider incentive payments using<br />

ACIR data.<br />

102 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


<strong>Immunisation</strong> service providers enrolled on VIVAS have access to a range<br />

of services, such as reminder notices for overdue or unimmunised patients,<br />

individual vaccination records via local public health units, and Queensland’s<br />

centralised Vaccine Distribution System (www.health.qld.gov.au/immunisation/<br />

health_professionals/vivas.asp).<br />

<strong>The</strong> Northern Territory<br />

<strong>The</strong> NT <strong>Immunisation</strong> Register records details of all vaccines administered<br />

to anyone in the Northern Territory (NT). <strong>Immunisation</strong> service providers<br />

in the NT are encouraged to report all administered vaccines to the NT<br />

<strong>Immunisation</strong> Register. This can be done by direct electronic transfer from<br />

some services or via printed lists from clinical software programs and/or by<br />

completing the NT childhood or adult vaccination recording form<br />

(www.health.nt.gov.au/Centre_for_Disease_Control/<strong>Immunisation</strong>/<br />

Recording_and_Reporting_Forms/index.aspx).<br />

<strong>The</strong> NT <strong>Immunisation</strong> Register routinely provides relevant data on vaccination<br />

encounters to the ACIR and the HPV Register.<br />

<strong>The</strong> NT <strong>Immunisation</strong> Register provides a number of services, such as recall lists<br />

for childhood immunisations in remote areas, individual vaccination records,<br />

and web-based access for NT immunisation service providers for immunisation<br />

histories for children


PART 3 VACCINATION FOR SPECIAL RISK GROUPS<br />

3.1 VACCINATION FOR ABORIGINAL AND<br />

TORRES STRAIT ISLANDER PEOPLE<br />

After European colonisation, Aboriginal and Torres Strait Islander (Indigenous)<br />

peoples experienced successive epidemics of infectious diseases with very<br />

high morbidity and mortality; many of these diseases have now become<br />

vaccine preventable. <strong>The</strong> diseases with the most serious impact were smallpox,<br />

tuberculosis, influenza, measles and syphilis, with estimated mortality rates of<br />

over 30% for smallpox epidemics and 20% for measles epidemics. 1 <strong>The</strong>se high<br />

rates of disease were mainly due to a lack of previous exposure, 2 followed by<br />

high-density living in newly established settlements. Over many decades, higher<br />

rates of infectious disease have been associated with lower standards of living<br />

and poorer access to water, housing and health care. 3 Social determinants of<br />

health, such as low educational outcomes, lack of control over life circumstances<br />

and lack of cultural safety, are also associated with poor health outcomes,<br />

including increased infectious disease risk. 4<br />

In recent decades, vaccination has been very successful in eliminating or<br />

substantially reducing the rates of many vaccine-preventable diseases (VPDs),<br />

such as diphtheria, polio, tetanus, hepatitis B, measles, mumps and rubella, in<br />

all <strong>Australian</strong>s, and has made a substantial contribution to improvements in<br />

Indigenous child mortality. 5 For some VPDs, control is suboptimal in the general<br />

population despite high vaccination coverage (e.g. pertussis). For others, such<br />

as invasive pneumococcal disease (IPD), greater burdens of illness still occur in<br />

Indigenous persons than in non-Indigenous persons, largely due to the greater<br />

prevalence in Indigenous persons of serotypes for which vaccines do not protect,<br />

and high exposure levels associated with the environmental issues mentioned<br />

above. 5 Timeliness of immunisation can also be a factor. 5<br />

In recognition of the higher rates of disease in the Indigenous population, some<br />

vaccines are specifically recommended for use in Indigenous persons, or for<br />

administration to a broader age range than is recommended for non-Indigenous<br />

persons (see Table 3.1.1). This chapter discusses the vaccines for which there are<br />

currently different recommendations for Indigenous persons in at least some<br />

parts of Australia, or for which there have been recent changes in this respect.<br />

For children, these are bacille Calmette-Guérin (BCG), Haemophilus influenzae<br />

type b, hepatitis A, influenza and pneumococcal vaccines. For adults, these are<br />

hepatitis B, influenza and pneumococcal polysaccharide vaccines.<br />

104 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


PART 3 VACCINATION FOR SPECIAL RISK GROUPS 105<br />

Table 3.1.1: Additional* vaccines recommended for Indigenous persons, due to their higher risk of disease<br />

Vaccine Recommendation for Indigenous persons<br />

BCG Neonates living in areas of high TB incidence †<br />

1 dose<br />

Hepatitis A Children resident in the Northern Territory, Queensland, South Australia and Western<br />

Australia<br />

Hepatitis B<br />

2 doses in the 2nd year of life‡<br />

Adults who have not previously been vaccinated against hepatitis B and are non-immune<br />

Influenza All persons aged ≥15 years<br />

Consider in all children aged ≥6 months, especially those aged


3.1.1 Children<br />

BCG vaccine and tuberculosis<br />

BCG vaccine is recommended for Indigenous neonates in regions of high<br />

tuberculosis (TB) incidence, where infants are at higher risk of acquiring this<br />

serious condition. BCG vaccine is provided for Indigenous neonates in the<br />

Northern Territory, Queensland and parts of northern South Australia, but<br />

no longer in Western Australia. 6 State/territory health authorities should be<br />

consulted to determine the recommendations for particular areas, including<br />

where BCG vaccination is being considered for neonates


Thus, a vaccine to prevent Hib disease in Indigenous children needed to be<br />

immunogenic as early as possible in infancy. <strong>The</strong> previously used Hib-containing<br />

vaccines, known by the abbreviation PRP-OMP, were more immunogenic at<br />

2 months of age than the other conjugate Hib (PRP-T) vaccines, and so were the<br />

preferred Hib vaccine type for Indigenous children in the first Hib vaccination<br />

programs beginning in 1993. Since then, there has been a dramatic decline in<br />

Hib disease in Indigenous children. 14,15 New combination vaccines that include<br />

a Hib (PRP-T) component, and have the advantage of reducing the number of<br />

injections, were introduced in some jurisdictions from November 2005. Initially<br />

PRP-T vaccines were not recommended for Indigenous children in jurisdictions<br />

with higher disease incidence, but either PRP-OMP or PRP-T vaccine could<br />

be given to other children. Following an international shortage of PRP-OMP<br />

vaccine, it was progressively replaced by PRP-T-containing vaccines for all<br />

children. Invasive Hib disease and nasopharyngeal colonisation with Hib are<br />

being closely monitored in high-incidence settings such as the Northern Territory<br />

and Western Australia following this change. To date, there has not been any<br />

change in Hib epidemiology found in association with the change to PRP-Tcontaining<br />

vaccines for Indigenous children.<br />

Hepatitis A<br />

Hepatitis A vaccination is recommended for Indigenous children in those<br />

jurisdictions with high disease incidence: the Northern Territory, Queensland,<br />

South Australia and Western Australia (see 4.4 Hepatitis A). Two doses should<br />

be given, commencing in the 2nd year of life. <strong>The</strong> recommended ages of<br />

administration vary between states and territories, so jurisdictional health<br />

authorities should be contacted for further details about local vaccination<br />

schedules.<br />

Hepatitis A infection was common during the 1990s in Indigenous children<br />

across northern and central Australia. 16-18 Most children acquired the virus in the<br />

first 5 years of life, which is a typical finding in populations with disadvantaged<br />

living conditions. Although the symptoms of infection in early childhood<br />

are usually mild or absent, cases complicated by liver failure and death have<br />

been reported among Indigenous children in Far North Queensland18 and the<br />

Kimberley, 16 and recorded hospitalisation rates are more than 50 times higher in<br />

Indigenous children than in non-Indigenous children. 5 A vaccination program for<br />

Indigenous children was introduced in north Queensland in 1999 and resulted<br />

in a 92% decrease in the number of reported cases, from 787 cases in all children<br />

during the period 1996–1999 to 66 cases in the period 2000–2003. This decrease<br />

in hepatitis A disease was observed in both Indigenous and non-Indigenous<br />

children, suggesting a substantial herd immunity effect. 19 From 2005, the<br />

hepatitis A vaccination program was extended to include all Indigenous children<br />

aged ≤2 years resident in the Northern Territory, Queensland, South Australia<br />

and Western Australia. Notifications have fallen by over 90%, from more than<br />

50 per 100 000 in 2005 to less than 5 per 100 000 in 2009.<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 107<br />

3.1 VACCINATION FOR<br />

ABORIGINAL AND TORRES<br />

STRAIT ISLANDER PEOPLE


Hepatitis B<br />

See ‘Hepatitis B’ under ‘Adults’ below.<br />

Influenza<br />

Annual influenza vaccination is recommended for any person ≥6 months of age<br />

(see 4.7 Influenza) for whom it is desired to reduce the likelihood of becoming<br />

ill with influenza. Young infants and children aged


3.1.2 Adults<br />

Hepatitis B<br />

Indigenous persons should have their risks and vaccination status for<br />

hepatitis B reviewed, be offered testing for previous hepatitis B infection, and<br />

be offered vaccination if non-immune. (See also 4.5 Hepatitis B.)<br />

High rates of mortality and morbidity from hepatitis B among Indigenous<br />

persons have been recognised ever since the original identification of the<br />

‘Australia antigen’ in 1965. 29 Prior to vaccination, estimates of the prevalence<br />

of markers of previous infection in Indigenous communities ranged from 20<br />

to 100%. In the Northern Territory, the incidence of primary hepatocellular<br />

carcinoma was 10 times greater in Indigenous persons than in non-Indigenous<br />

persons, and comparable to high-incidence countries such as China. 30 Vaccination<br />

programs have had substantial impacts on infection and carriage rates in both<br />

Indigenous and non-Indigenous <strong>Australian</strong>s. 5,31 However, there is evidence that<br />

new infections continue to occur at a higher rate in Indigenous persons, 5,32-34<br />

probably due to a combination of pre-existing high carriage rates, susceptible<br />

persons in older age groups (who were not eligible for vaccination programs), 33,34<br />

low coverage in early targeted programs, 33,34 and a poorer immunological<br />

response to vaccination. 35<br />

Influenza<br />

Annual influenza vaccination is recommended for all Indigenous persons aged<br />

≥15 years (see 4.7 Influenza).<br />

Influenza and its complications, especially secondary pneumonia, have<br />

historically been major causes of morbidity and mortality in Indigenous persons,<br />

both during and outside pandemic periods. This is probably related to a high<br />

prevalence of medical risk factors such as diabetes, renal disease and excessive<br />

alcohol use, as well as poorer environmental living conditions that facilitate<br />

disease transmission. 3 Past pandemics have had disproportionately severe<br />

impacts on Indigenous persons, 1 as did the influenza A(H1N1)pdm09 pandemic<br />

in 2009. Reported rates of infection, hospitalisation and death due to pandemic<br />

A(H1N1)pdm09 were 6.6, 6.2 and 5.2 times higher, respectively, in Indigenous<br />

persons than in non-Indigenous persons. 36<br />

In recent times, respiratory disease has been responsible for around 8% of deaths<br />

in Indigenous persons, the vast majority being in adults. 3 Influenza disease<br />

incidence is greatest in young children and the elderly. Hospitalisation rates<br />

due to influenza and pneumonia are highest in young children and lowest in<br />

older children. Hospitalisation and death rates increase with age in all adults,<br />

but increase much earlier in Indigenous adults than in non-Indigenous adults.<br />

<strong>The</strong> vast majority of these hospitalisations and deaths are due to pneumonia,<br />

but it is not clear what proportion of these are associated with influenza. 5<br />

Younger Indigenous adults (aged 25–49 years) have influenza and pneumonia<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 109<br />

3.1 VACCINATION FOR<br />

ABORIGINAL AND TORRES<br />

STRAIT ISLANDER PEOPLE


disease rates similar to older non-Indigenous adults (aged ≥50 years), but have<br />

much higher rates than non-Indigenous persons of the same age. In younger<br />

Indigenous adults rates are at least 8 times higher for hospitalisations and<br />

20 times higher for deaths, compared to younger non-Indigenous adults. 5 In<br />

addition, hospitalisation rates in Indigenous children aged


een low in younger Indigenous adults, an issue that requires attention if the<br />

full benefits of vaccination are to be realised. 5<br />

Other diseases<br />

Japanese encephalitis<br />

<strong>The</strong> first ever outbreak of Japanese encephalitis (JE) in Australia occurred in the<br />

remote outer islands of the Torres Strait in 1995, with 3 cases, 2 of them fatal.<br />

<strong>The</strong>re have been 5 cases to date acquired in Australia. Since then, JE virus has<br />

been detected frequently in sentinel animal surveillance in the outer islands.<br />

However, the sentinel pig surveillance system has been gradually disbanded<br />

since 2006, with surveillance of the last remaining herd on Cape York ceasing<br />

from the 2011–2012 wet season.<br />

A JE vaccine (JE-Vax) was first offered to the residents of the Torres Strait Islands<br />

in late 1995 and the vaccine was integrated into the vaccination schedule for<br />

children resident in the Torres Strait Islands, commencing at 12 months of age. 43,44<br />

<strong>The</strong>re has not been a case of JE in the Torres Strait since 1998 and the risk of<br />

JE has diminished considerably in the outer islands since the mid-1990s. Most<br />

communities have relocated pigs well away from homes, and major drainage<br />

works on most islands have markedly reduced potential breeding sites for<br />

vector mosquitoes.<br />

In 2007, the supply of JE-Vax vaccine into Australia ceased as the manufacturer<br />

stopped production. Because of this shortage of vaccine, for a short period from<br />

September 2007 JE-Vax was restricted to use on the six outer Torres Strait Islands:<br />

Badu, Boigu, Dauan, Mabuiag, Moa and Sabai. Two new JE vaccines, Imojev<br />

and JEspect, are now available for use in those at risk in the Torres Strait<br />

(see 4.8 Japanese encephalitis).<br />

Rubella<br />

Although evidence suggests that endemic rubella is well controlled in Australia,<br />

Indigenous women living in rural and remote regions are more likely to be nonimmune<br />

to rubella than non-Indigenous non-overseas-born <strong>Australian</strong>s. 45 Every<br />

effort should be made to identify non-pregnant seronegative Indigenous women<br />

of child-bearing age and provide measles-mumps-rubella (MMR) vaccine, in<br />

order to prevent congenital rubella syndrome (see 4.18 Rubella). Vaccination with<br />

MMR vaccine also ensures adequate protection against measles (see 4.9 Measles).<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 111<br />

3.1 VACCINATION FOR<br />

ABORIGINAL AND TORRES<br />

STRAIT ISLANDER PEOPLE


3.1.3 Service delivery<br />

General practitioners, Aboriginal Community Controlled Health Services,<br />

Community Health Services, the Royal Flying Doctor Service and state/territory<br />

corrective services all provide vaccination services to Indigenous persons and<br />

are important to the success of programs to vaccinate Indigenous persons. While<br />

vaccination coverage estimates vary over time and between communities, a<br />

relatively consistent finding has been higher coverage in Indigenous persons in<br />

remote areas than in urban areas. 46,47 More recently, however, this has not been<br />

the case for Indigenous children, where coverage has been high in both remote<br />

and urban areas; 48 coverage in remote areas is lower for adults than for children. 5<br />

For vaccines recommended for both Indigenous and non-Indigenous persons,<br />

coverage is as high, or higher, in Indigenous persons as in non-Indigenous<br />

persons, 5 but vaccination is more frequently delayed. 49-52 For example, one study<br />

reported that at 7 months of age only 45.2% of Indigenous infants in the Northern<br />

Territory had completed the recommended schedule for that age point (DTPahepB-IPV-Hib/PCV/rotavirus),<br />

but by 18 months of age this figure had risen<br />

to 81.2%. 51 Coverage for vaccines recommended only for Indigenous persons is<br />

generally lower than for vaccines that are funded for all persons in a particular<br />

age group. 53<br />

<strong>The</strong>se disparities point to the importance of identification of Indigenous status,<br />

particularly in mainstream health services, and particularly in urban areas. <strong>The</strong><br />

use of patient information systems to record Indigenous status and schedule<br />

preventive health services has the potential to increase opportunistic vaccination<br />

and enable the provision of patient reminders, with resultant improvements<br />

in coverage and timeliness. 54 Culturally appropriate service delivery and<br />

communication strategies, as well as use of Indigenous-specific Medicare<br />

items, will also assist in improving access to health services for Indigenous<br />

<strong>Australian</strong>s. 55-57<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

112 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


3.2 VACCINATION FOR INTERNATIONAL TRAVEL<br />

3.2.1 Introduction<br />

<strong>The</strong> number of <strong>Australian</strong>s who travel overseas has increased over recent years.<br />

Data available through the <strong>Australian</strong> Bureau of Statistics suggest that there were<br />

about 6.7 million short-term departures in 2010, with more than half travelling to<br />

destinations other than New Zealand or countries in North America and Europe. 1<br />

<strong>The</strong>re are various risks to health associated with international travel, including<br />

exposures to infective agents, extremes of altitude and temperature, and other<br />

physical, psychological and environmental hazards. <strong>The</strong>re could also be poor<br />

quality or limited access to clean water, shelter, hygiene and sanitation facilities,<br />

and health and medical care. <strong>The</strong> level of health risks will vary with individual<br />

factors, including the travellers’ underlying health and physiological state, the<br />

itinerary and activities undertaken, and the duration of exposure to various<br />

hazards during travel.<br />

Travellers with increased risks to their health include young children and<br />

infants; pregnant women; people with underlying medical conditions, especially<br />

immunocompromising conditions due to disease or medical treatment; travellers<br />

spending extended periods in multiple regions with poor resources or in remote<br />

regions; those participating in mass gatherings (major sporting, cultural, social<br />

or religious events involving large numbers of people); and migrant families<br />

travelling back to their country/region of origin to visit friends and relatives<br />

(VFR). Those undertaking VFR travel are more likely to have closer contact<br />

with local populations, stay in remote or rural areas, and consume higher-risk<br />

food and beverages. <strong>The</strong>y are also less likely to adequately perceive health<br />

risks associated with travelling, specifically seek pre-travel health advice, or be<br />

adequately vaccinated or prophylaxed. 2,3<br />

3.2.2 Infections acquired by travellers<br />

Exposure to infectious diseases, some of which are vaccine preventable, is one of<br />

the many health hazards of international travel. Although some of these diseases<br />

are present in Australia, the risk of acquiring them overseas may be higher<br />

because of higher disease incidence in other countries and/or increased risk of<br />

exposure resulting from activities undertaken during the travel period.<br />

Common infections acquired by travellers include those that follow ingestion<br />

of contaminated food or beverages. 4,5 Most of these are diarrhoeal diseases due<br />

to enteric pathogens, but infections with extra-intestinal manifestations, such as<br />

hepatitis A and typhoid, are also acquired this way. Vaccines against hepatitis A,<br />

typhoid and cholera are available.<br />

Insect-borne (particularly mosquito-borne) infections, such as malaria and<br />

dengue, are important causes of fever in <strong>Australian</strong> travellers returning from<br />

endemic areas, particularly Southeast Asia and Oceania. 5 Japanese encephalitis<br />

occurs throughout a large part of Asia and the Western Pacific region, including<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 113<br />

3.2 VACCINATION FOR<br />

INTERNATIONAL TRAVEL


Papua New Guinea. Yellow fever occurs only in parts of Africa and Central and<br />

South America, while tick-borne encephalitis occurs in parts of Europe and Asia.<br />

Vaccines are available for protection against Japanese encephalitis, yellow fever<br />

and tick-borne encephalitis.<br />

Vaccine-preventable infections transmitted via aerosols and/or droplets include<br />

influenza, meningococcal disease, measles, mumps and varicella (chickenpox);<br />

influenza is typically the most frequent vaccine-preventable infection among<br />

travellers. 6 Incidences of measles and mumps are higher in many overseas<br />

countries, including some developed countries, than in Australia. Tuberculosis is<br />

a rare infection in travellers, and is more likely to be acquired by expatriates who<br />

live in endemic areas for long periods than by short-term visitors.<br />

Blood-borne and sexually transmitted infections, such as hepatitis B, hepatitis C<br />

and human immunodeficiency virus (HIV), may pose a threat to some <strong>Australian</strong><br />

travellers. In some areas, there is the possibility that these viruses and other<br />

blood-borne agents may be transmitted by healthcare workers using non-sterile<br />

medical equipment or other poor infection control practices. Hepatitis B vaccine<br />

is relevant to many travellers.<br />

Travellers may be exposed to a variety of other exotic infectious agents, such<br />

as rabies (from bites or scratches from rabid dogs and other mammals in many<br />

countries), schistosomiasis (from exposure to water infested with the parasites, in<br />

Africa in particular), and leptospirosis (through activities like rafting or wading<br />

in contaminated streams). Of these, only rabies can be prevented by vaccination.<br />

Some other vector-borne diseases and parasitic (including protozoal and<br />

helminthic) diseases are also important for international travellers, some<br />

of which are preventable through appropriate barrier precautions and<br />

chemoprophylaxis (e.g. malaria).<br />

3.2.3 Practical aspects of recommending vaccinations for travellers<br />

Although important, recommending appropriate vaccinations is not the only<br />

component of a pre-travel medical consultation, and vaccines relevant for<br />

travelling are not restricted to those for prevention of diseases that occur most<br />

commonly overseas (‘travel vaccines’). Recommendation of a vaccine for<br />

travelling only on the basis of the destination country is undesirable. <strong>The</strong>re is no<br />

single ‘correct’ list of vaccines for travelling to any single country.<br />

In a pre-travel medical consultation, it is prudent to also acquire adequate<br />

information regarding:<br />

• relevant personal information of the traveller, including age, pregnancy<br />

or planning of pregnancy, or even possible financial constraints<br />

• underlying medical conditions of the traveller, particularly<br />

immunocompromising conditions, and current medications<br />

114 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


• vaccination history (including adverse events following immunisation) and<br />

allergy history of the traveller<br />

• detailed intended itinerary, including date of departure (and time period<br />

available for vaccinations), specific localities and routes, rural versus urban<br />

stay, duration of stay, likely access to healthcare and other services, and<br />

probability of deviation from planned itinerary<br />

• purpose(s) of travel and intended activities, especially those susceptible to<br />

various environmental risks and hazards<br />

• plans for travel insurance.<br />

This information will not only facilitate recommendations of preventive<br />

vaccinations and/or chemoprophylaxis that are commensurate with exposure<br />

risks and tailored to the proposed trip, but also provision of other important<br />

preventive health advice (e.g. food and water precautions, avoidance of bites<br />

from mosquitoes or other arthropods) and advice regarding management of<br />

possible health conditions during travel.<br />

Some overseas organisations, such as schools, colleges and universities, have<br />

policies requiring evidence of vaccination and/or immunity against some<br />

vaccine-preventable diseases, for example, measles and meningococcal disease.<br />

<strong>The</strong>se requirements should be taken into account while planning and scheduling<br />

immunisation prior to departure.<br />

<strong>The</strong> vaccination needs for a traveller may be conveniently considered in<br />

several categories.<br />

Routinely recommended vaccines (not specifically related to travelling overseas)<br />

All travellers should be up to date with current standard vaccination<br />

recommendations. Consideration should also be given to any other vaccines<br />

that may be relevant to the individual’s health status or underlying medical<br />

conditions, occupation or lifestyle (e.g. pneumococcal polysaccharide vaccine for<br />

an elderly person or person otherwise recommended to have had pneumococcal<br />

vaccine, hepatitis B vaccine for a first aid officer). <strong>The</strong> probability of exposure<br />

to some of these diseases may be greater while travelling overseas, even to<br />

‘developed’ countries (e.g. measles and mumps). For some itineraries, it may be<br />

appropriate for some booster doses to be received sooner (i.e. before travel) than<br />

at the routine recommended time (e.g. diphtheria-tetanus booster).<br />

Selected vaccines based on travel itinerary, activities and likely risk of<br />

disease exposure<br />

A risk assessment approach should be adopted in recommending some selective<br />

vaccines based on travel itineraries (‘travel vaccines’). Potential risks of disease<br />

exposure and protective benefits from vaccinations should be weighed against<br />

potential adverse effects and both non-financial and financial costs arising from<br />

vaccinations. Priority should be given to vaccines for diseases that are common<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 115<br />

3.2 VACCINATION FOR<br />

INTERNATIONAL TRAVEL


and of significant impact (e.g. influenza and hepatitis A), and to those diseases<br />

that, although less common, have severe potential adverse outcomes (e.g.<br />

Japanese encephalitis and rabies). Booster doses should be considered where<br />

appropriate (see Table 3.2.1). Because of the imminence of departure, sometimes<br />

an ‘accelerated schedule’ may be considered appropriate (e.g. for hepatitis B or<br />

the combined hepatitis A/hepatitis B vaccine – see the relevant disease-specific<br />

chapters in Part 4). Note that, while immunity may be established sooner with<br />

the accelerated schedule, an additional dose is required about a year later for<br />

completion of the course to augment long-term protection. For children, the<br />

lower age limits for recommendation of selected vaccines should be noted (see<br />

Table 3.2.2).<br />

Vaccines required by International Health Regulations or for entry into<br />

specific countries<br />

Currently, yellow fever vaccination is the only vaccination that may be required<br />

by the International Health Regulations for travelling in certain situations, for<br />

the purpose of individual protection if one is likely to be exposed and/or for<br />

protection of vulnerable populations (in countries with relevant vectors) from<br />

importation of the disease. Some countries, including Australia, may require<br />

documented evidence of yellow fever vaccination as a condition of entry or exit.<br />

Although there are exceptions, this would mostly be relevant for travellers who<br />

originate from, or have travelled or made transit through, countries in Africa or<br />

South America (see 4.23 Yellow fever and 3.2.6 Further information below). Current<br />

requirements should be referred to when advising travellers regarding yellow fever<br />

immunisation requirements.<br />

<strong>The</strong> Ministry of Health of Saudi Arabia annually issues specific requirements<br />

and recommendations for entry visas for travellers on pilgrimage to Mecca in<br />

Saudi Arabia (Hajj and Umra). For pilgrims travelling directly from Australia,<br />

only evidence of quadrivalent meningococcal vaccination is currently mandatory.<br />

However, current requirements should be referred to when advising prospective<br />

Hajj and Umra pilgrims (see 3.2.6 Further information below).<br />

Vaccine administration and documentation<br />

It is not unusual that multiple vaccines would be required before travelling.<br />

<strong>The</strong> standard recommendations and precautions for administration of multiple<br />

vaccines are applicable (see 2.2 Administration of vaccines).<br />

Multiple clinic visits may be necessary when multiple vaccines and vaccines<br />

that involve multiple doses are involved (e.g. rabies pre-exposure prophylaxis<br />

or hepatitis B vaccine). Special attention should be paid to the appropriate<br />

scheduling of these visits, taking into account dose interval precautions (e.g.<br />

multiple live vaccines), requirement for pre-vaccination tests (e.g. tuberculin<br />

skin test), and potential interference by some antimalarials if relevant (e.g. rabies<br />

vaccine). Ideally, vaccination courses should be started early enough before<br />

116 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


departure to allow for the period when most adverse events are expected to<br />

occur and to allow sufficient time for adequate immunity to develop.<br />

It is important to document travel vaccines appropriately, not only in the<br />

clinic’s record but also in a suitable record that can be carried by the traveller.<br />

It is recommended that the record also includes all the other ‘routinely<br />

recommended’ vaccines that the traveller has ever received. An International<br />

Certificate of Vaccination or Prophylaxis (ICVP), issued by an authorised medical<br />

practitioner in accordance with the International Health Regulations (2005), is<br />

required for yellow fever vaccination.<br />

3.2.4 Vaccines<br />

Detailed information regarding each of the vaccines discussed below is provided<br />

in each of the corresponding disease-specific chapters of this <strong>Handbook</strong>. This<br />

section provides some general guidance in considering whether a particular<br />

vaccine may be advisable for a traveller.<br />

All prospective travellers should have been vaccinated according to the<br />

recommended vaccination schedule appropriate for the traveller’s age and<br />

underlying health conditions. All children should be vaccinated according<br />

to the NIP schedule. In exceptional circumstances, the NIP vaccines may be<br />

administered at the minimum age rather than the recommended age (see 2.1.5<br />

Catch-up, Table 2.1.5 Minimum acceptable age for the 1st dose of scheduled vaccines<br />

in infants in special circumstances). Children vaccinated using the minimum age<br />

rather than the recommended age may require extra vaccine doses to ensure<br />

adequate protection. <strong>The</strong> minimum interval requirements between doses must<br />

be observed (see 2.1.5 Catch-up, Table 2.1.7 Minimum acceptable dose intervals for<br />

children


of intermediate or high endemicity of hepatitis B, including Central and South<br />

America, Africa, Asia or Oceania, are recommended to be vaccinated against<br />

hepatitis B, due to the potential for inadvertent exposure to hepatitis B virus<br />

through blood-borne or sexual routes, including unplanned medical or dental<br />

procedures. A survey has shown that about half of <strong>Australian</strong> travellers who<br />

spent at least 3 nights in Southeast or East Asia had participated in at least one<br />

activity with a risk of acquiring hepatitis B. 7 (See also 4.5 Hepatitis B.)<br />

Influenza and pneumococcal disease<br />

Older travellers (usually those aged ≥65 years) and those with any relevant<br />

underlying medical or behavioural risk factors (see 4.7 Influenza and 4.13<br />

Pneumococcal disease) should receive the seasonal influenza vaccine and/or<br />

should have received the 23-valent pneumococcal polysaccharide vaccine. All<br />

travellers should consider influenza vaccine, especially if travelling during the<br />

influenza season of the destination region(s). <strong>The</strong> influenza vaccine is particularly<br />

relevant if influenza epidemics are occurring at the traveller’s destination(s), and<br />

for travellers in large tourist groups, especially those that include older persons,<br />

or travelling on cruises, where they are likely to be in confined circumstances for<br />

days to weeks (see 4.7 Influenza).<br />

Measles, mumps, rubella and varicella<br />

Most measles outbreaks in Australia now result from an infection imported by<br />

inadequately vaccinated young travellers. Incidences of measles and mumps are<br />

higher in some overseas countries, regions or communities, including developed<br />

countries, than in Australia. <strong>Australian</strong>s born during or since 1966 who have<br />

not received 2 doses of measles-, mumps- and rubella-containing vaccines<br />

should be vaccinated with the MMR vaccine before travelling (noting pregnancy<br />

precautions) (see 4.9 Measles). Varicella vaccine should be offered to unvaccinated<br />

travellers who have not had clinical disease, or where serology demonstrates<br />

lack of immunity in those with an uncertain history of clinical disease (see 4.22<br />

Varicella).<br />

Poliomyelitis<br />

All travellers should be age-appropriately immunised against polio (see 4.14<br />

Poliomyelitis). If travelling to countries where wild poliovirus transmission still<br />

occurs (i.e. Afghanistan, Pakistan, Nigeria and others where polio may have<br />

been re-established or have been imported – check recent updates), inactivated<br />

poliomyelitis vaccine (IPV) should be offered to those who have not completed a<br />

3-dose primary course of any polio vaccine, and a single booster dose should be<br />

given to those who have previously completed the primary course. For an up-todate<br />

list of affected countries see www.polioeradication.org.<br />

118 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Selected vaccines based on travel itinerary, activities and likely risk of<br />

disease exposure<br />

Cholera<br />

Cholera vaccination is rarely indicated for most travellers, 8 as the risk of<br />

acquiring cholera for travellers in general is very low, provided that general<br />

precautions to avoid contaminated food and water are taken. <strong>The</strong> protective<br />

efficacy against Vibrio cholerae O1 is high (>80%) among children aged 2–5<br />

years for the initial 4–6 months after 3 doses, but wanes to become insignificant<br />

afterwards. For those aged >5 years, protective efficacy is about 78% and 63% for<br />

the 1st and 2nd year, respectively, and wanes to become insignificant beyond 2<br />

years after vaccination. 9 <strong>The</strong> vaccine does not protect against the V. cholerae O139<br />

serogroup. It is only indicated for those travellers at considerable risk, such as<br />

those working in humanitarian disaster situations. However, since cholera and<br />

enterotoxigenic Escherichia coli (ETEC) share the same toxin, cholera vaccination<br />

does afford some partial short-term protection against ETEC-caused travellers’<br />

diarrhoea. <strong>The</strong> effect lasts only about 3 months, and the overall reduction of<br />

travellers’ diarrhoea risk would be less than 15%; 10 however, there may be some<br />

travellers who would benefit from improved protection against travellers’<br />

diarrhoea, including those with achlorhydria and those at increased risk of severe<br />

or complicated diarrhoeal disease (see 4.1 Cholera).<br />

Certification of cholera vaccination has been abandoned globally, and no<br />

countries have official entry requirements for cholera vaccination.<br />

Hepatitis A<br />

Hepatitis A vaccine should be recommended to all travellers ≥1 year of age<br />

travelling to moderately or highly endemic countries (including all developing<br />

countries), except those who are likely to have acquired natural immunity<br />

following previous infection (see 4.4 Hepatitis A). <strong>The</strong>re is no longer any place<br />

for the routine use of normal human immunoglobulin to prevent hepatitis A in<br />

travellers (see 4.4 Hepatitis A).<br />

Japanese encephalitis<br />

<strong>The</strong> mosquito-borne Japanese encephalitis virus is endemic in many countries<br />

in Asia and the Western Pacific region, including Papua New Guinea. High risk<br />

of transmission occurs in rural irrigated paddy and pig-farming environments.<br />

Vaccination is recommended for travellers spending a month or more in rural<br />

areas of high risk in endemic regions and should be considered for shorter-term<br />

travellers, particularly if travel is during the wet season or anticipated to be<br />

repeated, and/or there is considerable outdoor activity and/or accommodation<br />

is inadequately screened against mosquitoes. Vaccination is also recommended<br />

for expatriates spending a year or more in Asia, even if much of the stay is in<br />

urban areas (see 4.8 Japanese encephalitis).<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 119<br />

3.2 VACCINATION FOR<br />

INTERNATIONAL TRAVEL


Meningococcal disease<br />

All children ≥12 months of age receive meningococcal C conjugate vaccine<br />

(MenCCV) under the NIP.<br />

Up-to-date epidemiological information should be sought to determine the need<br />

for meningococcal vaccination in travellers. <strong>The</strong> quadrivalent meningococcal<br />

vaccine (which includes serogroups A, C, W and Y antigens) is recommended<br />

135<br />

for those who intend travelling to parts of the world where epidemics of<br />

meningococcal disease occur, in particular the ‘meningitis belt’ of sub-Saharan<br />

Africa. 11 <strong>The</strong> Saudi Arabian authorities require that all pilgrims travelling to<br />

Mecca (for the Hajj or Umra) have evidence of recent vaccination with the<br />

quadrivalent meningococcal vaccine8 (see 3.2.6 Further information below). <strong>The</strong><br />

quadrivalent meningococcal conjugate vaccine (4vMenCV) should be used in<br />

preference to the quadrivalent meningococcal polysaccharide vaccine (4vMenPV)<br />

(see 4.10 Meningococcal disease). <strong>The</strong>re is currently no vaccine available against<br />

serogroup B meningococcal disease.<br />

Rabies<br />

Travellers to rabies-endemic regions should be advised of the risk of rabies<br />

infection, and to avoid close contact with either wild, stray or domestic animals,<br />

in particular dogs, cats, monkeys and bats. Travellers should also be aware of<br />

the importance of appropriate immediate wound care of all animal bites and<br />

scratches (see 4.16 Rabies and other lyssaviruses (including <strong>Australian</strong> bat lyssavirus)).<br />

Recommendation for pre-travel (i.e. pre-exposure prophylaxis) rabies vaccination<br />

(or, where indicated, booster doses) is based on an assessment of the likelihood<br />

of contact and risk of exposure to potentially rabid animals, the access to<br />

appropriate healthcare and availability of post-exposure prophylaxis, including<br />

rabies immunoglobulin, should there be an at-risk exposure, and the timeliness<br />

of such access after exposure. <strong>The</strong> previous recommendation for pre-exposure<br />

prophylaxis based on duration of stay in rabies-endemic areas (i.e. for more<br />

than a month) is arbitrary, and most <strong>Australian</strong> travellers who have required<br />

post-exposure prophylaxis have undertaken shorter periods of travel. A lower<br />

threshold for recommending rabies pre-exposure prophylaxis should be adopted<br />

for children travelling to endemic areas (see 4.16 Rabies and other lyssaviruses<br />

(including <strong>Australian</strong> bat lyssavirus)). Vaccination against rabies before travel<br />

ensures that a safe and efficacious vaccine has been used and simplifies the<br />

management of a subsequent exposure because fewer doses of vaccine are<br />

needed. It also means that rabies immunoglobulin, which is often extremely<br />

expensive, difficult or even impossible to obtain in many developing countries, is<br />

not required, and reduces the urgency of post-exposure prophylaxis.<br />

120 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Tick-borne encephalitis<br />

Tick-borne encephalitis (TBE) is caused by a tick-borne RNA flavivirus and<br />

may involve the central nervous system. <strong>The</strong> disease is prevalent in parts of<br />

temperate regions of central and northern Europe and across northern Asia.<br />

Travellers are at particular risk when hiking or camping in forested areas in<br />

endemic regions during the summer months. Safe and effective vaccines are<br />

available. Vaccination is recommended only for individuals with a high risk<br />

of exposure. Two inactivated TBE vaccine formulations (from Austria and<br />

Germany) are available in Europe (based on the European subtype), and two<br />

other formulations, based on the Far Eastern subtypes, are available in Russia.<br />

<strong>The</strong>re is limited evidence that suggests the Austrian and German vaccines induce<br />

cross-protecting immunity against the Far Eastern and Siberian subtypes. 12 While<br />

the conventional schedule for completing the primary vaccination course takes<br />

9 to 12 months, accelerated schedules are available (see 3.2.6 Further information<br />

below). While no TBE vaccine is registered in Australia, a small stock of vaccine<br />

may be available in Australia for use under the Special Access Scheme.<br />

Tuberculosis<br />

Vaccination with BCG vaccine is generally recommended for tuberculinnegative<br />

children


Typhoid<br />

Typhoid vaccine may be recommended to travellers ≥2 years of age travelling<br />

to endemic regions, including the Indian subcontinent, most Southeast Asian<br />

countries and several South Pacific nations, including Papua New Guinea. This<br />

advice is also relevant for those travelling (back) to endemic regions to visit<br />

friends and relatives (VFR travel). Inactivated parenteral or live oral typhoid<br />

vaccine formulations are available (see 4.21 Typhoid).<br />

Yellow fever<br />

<strong>The</strong> World Health Organization no longer routinely reports on yellow fever<br />

‘infected areas’. <strong>The</strong> yellow fever vaccine is now recommended for all travellers<br />

aged ≥9 months to areas where there is evidence of persistent or periodic<br />

yellow fever virus transmission, or when required by the country for entry (see<br />

4.23 Yellow fever). 3 Countries requiring yellow fever vaccination for entry have<br />

legislation to do so in accordance with the International Health Regulations.<br />

Country requirements are subject to change at any time. Updates can be<br />

found at www.who.int/ith. To minimise the risk of yellow fever introduction,<br />

some countries, including those without current disease transmission such as<br />

Australia, may require proof of vaccination from travellers who have travelled<br />

to countries with risk of transmission. See also the <strong>Australian</strong> Government<br />

Department of Health and Ageing’s yellow fever fact sheet (www.health.gov.au/<br />

yellowfever).<br />

<strong>The</strong> risk of being infected with the yellow fever virus, country entry<br />

requirements, and individual factors like age, pregnancy and underlying<br />

medical conditions must be taken into account when considering yellow fever<br />

vaccination. Vaccination is generally not recommended when travelling to areas<br />

where there is low potential for yellow fever virus exposure (i.e. no human<br />

yellow fever cases ever reported and evidence to suggest only low levels of<br />

yellow fever virus transmission in the past). However, vaccination might be<br />

considered for a small subset of travellers to these areas who are at increased<br />

risk of exposure to mosquitoes or unable to avoid mosquito bites. 3 For people<br />

aged ≥60 years, the risk of adverse events associated with the vaccine should be<br />

weighed against the potential for disease exposure and benefit of yellow fever<br />

vaccination, as this age group has an increased risk of severe adverse events after<br />

primary yellow fever vaccination13 (see 4.23 Yellow fever).<br />

122 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


PART 3 VACCINATION FOR SPECIAL RISK GROUPS 123<br />

Table 3.2.1: Dose and routes of administration of commonly used vaccines in adult travellers (the lower age limit for the<br />

adult dosage varies with individual vaccines – please refer to the product information)<br />

Vaccine (adults) Brand name Dose (adults) Route Dosing intervals Duration of immunity and/or<br />

booster recommendations<br />

Routinely recommended vaccines (not specifically related to travelling overseas)<br />

Diphtheria-tetanus<br />

(dT)<br />

Diphtheria-tetanuspertussis<br />

(dTpa)<br />

Diphtheria-tetanuspertussis-inactivated<br />

poliomyelitis (dTpa-<br />

IPV)<br />

ADT Booster 0.5 mL IM A primary course is 3 doses<br />

of dT-containing vaccine,<br />

Boostrix<br />

0.5 mL IM<br />

given a minimum of 4 weeks<br />

apart; followed by booster<br />

or<br />

Adacel<br />

doses 10 and 20 years after.<br />

Boostrix-IPV<br />

or<br />

Adacel Polio<br />

0.5 mL IM<br />

Hepatitis B Engerix-B 1.0 mL IM 0, 1, 6 months or<br />

0, 1, 2, 12 months or<br />

0, 7, 21 days and 12 months*<br />

H-B-Vax II 1.0 mL IM 0, 1, 6 months<br />

Influenza (seasonal) Various 0.5 mL IM or<br />

intradermal,<br />

depending on the<br />

formulation<br />

3.2 VACCINATION FOR<br />

INTERNATIONAL TRAVEL<br />

Prior to travel, adults should receive<br />

a booster dose of dT (or dTpa if not<br />

given previously), if more than 10<br />

years have elapsed since their last<br />

dose of dT-containing vaccine.<br />

For persons undertaking high-risk<br />

travel, consider giving a booster<br />

dose of either dTpa or dT (as<br />

appropriate) if more than 5 years<br />

have elapsed since their last dose of<br />

dT-containing vaccine.<br />

A completed series probably gives<br />

life-long immunity.<br />

Single dose As different strains circulate from<br />

year to year, annual vaccination<br />

with the current formulation is<br />

necessary.


124 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Vaccine (adults) Brand name Dose (adults) Route Dosing intervals Duration of immunity and/or<br />

booster recommendations<br />

Routinely recommended vaccines (not specifically related to travelling overseas)<br />

Measles-mumpsrubella<br />

Pneumococcal Prevenar 13<br />

or<br />

Pneumovax 23<br />

Priorix 0.5 mL SC/IM <strong>Australian</strong>s born during<br />

or since 1966 who do not<br />

have documented evidence<br />

of having received 2 doses<br />

of measles-, mumps- and<br />

M-M-R II 0.5 mL SC<br />

rubella-containing vaccine<br />

should receive at least 1<br />

dose of MMR vaccine before<br />

travel<br />

0.5 mL IM Single dose, for older<br />

adults, and younger adults<br />

with predisposing medical<br />

conditions – see 4.13<br />

Pneumococcal disease<br />

Poliomyelitis IPOL 0.5 mL SC For unvaccinated adults,<br />

3 doses with minimum<br />

interval of 1 to 2 months<br />

between doses<br />

Combination<br />

vaccines (dTpa-<br />

IPV)<br />

Varicella (chickenpox) Varilrix<br />

or<br />

Varivax<br />

Refrigerated<br />

A 2-dose schedule provides longlasting<br />

immunity.<br />

Recommendations vary according<br />

to age, Indigenous status and<br />

predisposing conditions – see 4.13<br />

Pneumococcal disease.<br />

A booster dose 10-yearly is<br />

only necessary if travelling to a<br />

poliomyelitis endemic country.<br />

See Diphtheria-tetanus-pertussis-inactivated poliomyelitis (dTpa-IPV) above and 4.14 Poliomyelitis.<br />

0.5 mL SC If there is a lack of reliable<br />

history of chickenpox or the<br />

person is non-immune, and<br />

has not been vaccinated in<br />

childhood<br />

0, 4 weeks if aged ≥14 years<br />

A 2-dose schedule provides longlasting<br />

immunity.


PART 3 VACCINATION FOR SPECIAL RISK GROUPS 125<br />

Vaccine (adults) Brand name Dose (adults) Route Dosing intervals Duration of immunity and/or<br />

booster recommendations<br />

Selected vaccines based on travel itinerary, activities and likely risk of disease exposure<br />

Hepatitis A Avaxim 0.5 mL IM 0, 6–12 months A completed series probably gives<br />

Havrix 1440 1.0 mL IM 0, 6–12 months<br />

life-long immunity.<br />

Hepatitis A/B<br />

combined<br />

Hepatitis A/typhoid<br />

combined<br />

Vaqta Adult<br />

formulation<br />

1.0 mL IM 0, 6–18 months<br />

Twinrix (720/20) 1.0 mL IM 0, 1, 6 months or<br />

0, 7, 21 days and 12 months*<br />

Vivaxim †<br />

Note: Only for use<br />

in persons ≥16<br />

years of age<br />

1.0 mL (mixed<br />

vaccine)<br />

3.2 VACCINATION FOR<br />

INTERNATIONAL TRAVEL<br />

A completed series probably gives<br />

life-long immunity to both hepatitis<br />

A and B.<br />

IM Single dose A dose of monovalent hepatitis A<br />

vaccine given 6–36 months later<br />

probably gives life-long immunity.<br />

<strong>The</strong> duration of protection against<br />

typhoid is probably 3 years.<br />

Japanese encephalitis JEspect 0.5 mL IM 0, 28 days <strong>The</strong> need for, and timing of, a<br />

booster dose has not yet been<br />

determined.<br />

Meningococcal<br />

ACW 135 Y<br />

(quadrivalent<br />

conjugate 4vMenCV)<br />

Imojev 0.5 mL SC Single dose <strong>The</strong> need for, and timing of, a<br />

booster dose has not yet been<br />

determined.<br />

Menveo<br />

or<br />

Menactra<br />

0.5 mL IM Single dose <strong>The</strong> need for, and timing of, a<br />

booster dose has not yet been<br />

determined. Where not otherwise<br />

indicated 4vMenCV should be<br />

given in preference to 4vMenPV.


126 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Vaccine (adults) Brand name Dose (adults) Route Dosing intervals Duration of immunity and/or<br />

booster recommendations<br />

Selected vaccines based on travel itinerary, activities and likely risk of disease exposure<br />

Meningococcal<br />

ACW 135 Y<br />

(quadrivalent<br />

polysaccharide<br />

4vMenPV)<br />

Rabies (pre-exposure<br />

prophylaxis)<br />

Mencevax ACWY<br />

or<br />

Menomune<br />

Mérieux<br />

Inactivated<br />

Rabies Vaccine<br />

Rabipur<br />

Inactivated<br />

Rabies Virus<br />

Vaccine<br />

Typhoid Vivotif Oral A single oral<br />

capsule per<br />

dose<br />

Typherix<br />

or<br />

Typhim Vi<br />

0.5 mL SC Single dose Where not otherwise indicated,<br />

4vMenCV should be given in<br />

preference to 4vMenPV.<br />

Give 3–5-yearly boosters if the<br />

person is at ongoing risk.<br />

1.0 mL IM/SC 0, 7, 21–28 days Boosters are not recommended for<br />

frequent travellers unless they are<br />

at ongoing, high occupational risk<br />

1.0 mL IM 0, 7, 21–28 days<br />

of exposure – then either measure<br />

rabies antibody titres (and boost if<br />

titres are reported as inadequate) or<br />

give a single booster dose 2-yearly.<br />

Oral One capsule each on days<br />

1, 3, 5 (3-dose course), and<br />

preferably also day 7 ‡ (4-dose<br />

course)<br />

If the person is at ongoing risk,<br />

repeat the course after 3 years if a<br />

3-dose course was given initially;<br />

repeat the course after 5 years if a<br />

4-dose course was given initially.<br />

0.5 mL IM Single dose Give 3-yearly boosters if the person<br />

is at ongoing risk.<br />

Yellow fever Stamaril 0.5 mL IM/SC Single dose Give 10-yearly boosters if the<br />

person is at ongoing risk.<br />

* This ‘rapid’ schedule should be used only if there is very limited time before departure to endemic regions.<br />

† Vivaxim is registered for use in persons aged ≥16 years.<br />

‡ A 4th capsule of oral typhoid vaccine on day 7 is preferred (see 4.21 Typhoid).


3.2.5 Vaccinating the traveller with special risk factors<br />

See 3.3 Groups with special vaccination requirements and the disease-specific<br />

chapters in Part 4 for recommendations for travellers who are either pregnant or<br />

immunocompromised.<br />

Children should receive relevant travel vaccines, according to age-specific<br />

dosage and schedules as shown in Table 3.2.2; further information relating to<br />

administration is provided in the relevant disease-specific chapters in Part 4.<br />

Particular effort should be made to encourage the families of recent migrants to<br />

Australia to seek health advice before travelling to their country of origin to visit<br />

relatives and friends. 14<br />

Table 3.2.2: Recommended lower age limits of travel vaccines for children*<br />

Vaccine Lower age limit Dose/route Dosing intervals<br />

Hepatitis A<br />

Avaxim<br />

Havrix Junior<br />

Vaqta Paediatric/Adolescent<br />

formulation<br />

Hepatitis A/B combined<br />

Twinrix Junior (360/10)<br />

Twinrix (720/20)<br />

Japanese encephalitis<br />

JEspect<br />

Imojev<br />

Meningococcal ACW 135 Y<br />

(quadrivalent conjugate<br />

4vMenCV)<br />

Menveo<br />

2 years<br />

2 years<br />

1 year<br />

1 year<br />

1 year<br />

1 year (to


Vaccine Lower age limit Dose/route Dosing intervals<br />

Rabies<br />

Pre-exposure:<br />

Mérieux Inactivated Rabies<br />

Vaccine<br />

Rabipur Inactivated Rabies<br />

Virus Vaccine<br />

Typhoid<br />

Vivotif Oral<br />

Typherix<br />

Typhim Vi<br />

Yellow fever<br />

No lower age limit<br />

No lower age limit<br />

6 years<br />

2 years<br />

2 years<br />

1.0 mL IM/<br />

SC<br />

1.0 mL IM<br />

Oral capsule<br />

0.5 mL IM<br />

0.5 mL IM<br />

Stamaril 9 months # 0.5 mL IM/<br />

SC<br />

128 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

3 doses: 0, 7, 21–28 days<br />

3 doses: 0, 7, 21–28 days<br />

One capsule each on days<br />

1, 3, 5 (3-dose course), and<br />

preferably also day 7 (4-dose<br />

course)<br />

Single dose<br />

Single dose<br />

Single dose<br />

* See also minimum ages in Table 2.1.5 Minimum acceptable age for the 1st dose of scheduled vaccines in<br />

infants in special circumstances.<br />

† This schedule is not recommended if prompt protection against hepatitis B is required<br />

(see 4.5 Hepatitis B).<br />

‡ JEspect is registered for use in persons aged ≥18 years, but can be administered to persons<br />

aged ≥12 months in circumstances where an alternative is not available or contraindicated<br />

(see 4.8 Japanese encephalitis).<br />

§ 4vMenCV is preferred over 4vMenPV (see 4.10 Meningococcal disease).<br />

A 4th capsule of oral typhoid vaccine on day 7 is preferred (see 4.21 Typhoid).<br />

# Yellow fever vaccine is contraindicated in infants


• Travel health and quarantine section of the <strong>Australian</strong> Government Department<br />

of Health and Ageing website (www.health.gov.au/internet/main/<br />

publishing.nsf/Content/health-pubhlth-strateg-quaranti-index.htm)<br />

• Smartraveller – the <strong>Australian</strong> Government’s travel advisory and consular<br />

information service, which provides up-to-date advice regarding health,<br />

safety and other risks of specific destinations to <strong>Australian</strong> travellers (www.<br />

smartraveller.gov.au).<br />

Comprehensive technical advice on international travel and health, including<br />

but not limited to vaccinations, is available in the latest editions of the WHO<br />

publication International travel and health (available at www.who.int/ith/en) and<br />

the US Centers for Disease Control and Prevention (CDC) publication Health<br />

information for international travel (the ‘Yellow book’) (available at www.cdc.gov/<br />

travel).<br />

<strong>The</strong> Ministry of Health of Saudi Arabia’s requirements and recommendations for<br />

travellers on pilgrimage to Mecca (Hajj and Umra) are published annually in the<br />

Weekly Epidemiological Record of the WHO (www.who.int/wer). 8<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 129<br />

3.2 VACCINATION FOR<br />

INTERNATIONAL TRAVEL


3.3 GROUPS WITH SPECIAL VACCINATION REQUIREMENTS<br />

This chapter considers the use of vaccines in persons who have special<br />

vaccination requirements, persons who may experience more frequent<br />

adverse events following immunisation and persons who may have a<br />

suboptimal response to vaccination. Recommendations for vaccination of<br />

persons at occupational or lifestyle-associated risk are also included. Although<br />

recommendations are discussed under each sub-heading in this chapter, it is<br />

also important to refer to the relevant disease-specific chapters in Part 4 for<br />

further information.<br />

Administration of certain vaccines is a priority for some persons with<br />

medical conditions that increase the risk of infectious diseases, even in the<br />

absence of specific immune defects, for example, the use of influenza and<br />

pneumococcal vaccines in individuals with an increased risk of complications<br />

from these diseases. <strong>The</strong> presence of additional recommendations specific to<br />

groups discussed in this section underpins the importance of pre-screening<br />

those attending for immunisation and being certain to regularly review the<br />

vaccination needs of those seeking medical attention for any reason.<br />

3.3.1 Vaccination of persons who have had an adverse event<br />

following immunisation<br />

Adverse reactions after being given a vaccine (also known as ‘vaccine side<br />

effects’) do sometimes occur. It is usually not possible to predict which<br />

individuals may have a mild or a rare, serious reaction to a vaccine. However, by<br />

following guidelines regarding when vaccines should and should not be used,<br />

the risk of adverse effects can be minimised. <strong>The</strong> term ‘adverse event following<br />

immunisation’ (AEFI) refers to any untoward medical occurrence that follows<br />

immunisation, whether expected or unexpected, and whether triggered by the<br />

vaccine or only coincidentally occurring after receipt of a vaccine dose. 1 For more<br />

information on AEFI, see 2.3.2 Adverse events following immunisation.<br />

Serious adverse events occur rarely after immunisation. Recognised rare<br />

and serious AEFI are described in 2.3.2 Adverse events following immunisation.<br />

Pre-vaccination screening should identify persons who have experienced an<br />

AEFI and also identify persons with conditions that are precautions and/or<br />

contraindications to vaccines (see Table 2.1.1 Pre-vaccination screening checklist).<br />

<strong>The</strong> relevant disease-specific chapter(s) in Part 4 of this <strong>Handbook</strong> should be<br />

consulted for each vaccine regarding contraindications and precautions that are<br />

relevant. In general, persons who have had a non-serious adverse event can be<br />

safely revaccinated by their usual immunisation service provider. Determining<br />

whether revaccination should be provided after a serious event has occurred<br />

following vaccination can be more challenging. At the individual patient<br />

level, an assessment should be made as to whether the vaccine(s) was causally<br />

related to the adverse event. This includes a thorough medical assessment,<br />

including determining the need for, or availability of, specific tests to predict<br />

130 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


whether the AEFI is likely to recur with subsequent doses. Persons who have<br />

experienced a serious adverse event following immunisation (other than a<br />

contraindication, such as anaphylaxis confirmed as triggered by a vaccine or one<br />

of its components) can usually subsequently be vaccinated under close medical<br />

supervision. However, further advice should be sought where appropriate,<br />

by referral to a specialist clinic for the management of persons with special<br />

vaccination requirements (including persons who have had a previous AEFI).<br />

Information about specialist immunisation clinics, or the contact details for<br />

paediatricians or medical specialists with experience in management of persons<br />

with AEFI, are usually available from state and territory health authorities (see<br />

Appendix 1 Contact details for <strong>Australian</strong>, state and territory government health<br />

authorities and communicable disease control) and from the Immunise Australia<br />

website (www.immunise.health.gov.au).<br />

Allergies<br />

Vaccines rarely produce allergy or anaphylaxis (a rapid and life-threatening<br />

form of allergic reaction). Overall, the risk of anaphylaxis after a single vaccine<br />

dose has been estimated as less than 1 case per 1 million; however, this risk<br />

varies depending on the vaccine type. 2 Antibiotics, gelatin and egg proteins<br />

are the components most often implicated in allergic reactions. Yeast has only<br />

rarely been associated with vaccine-related allergic reaction. Persons allergic to<br />

latex may be at risk from some vaccines. This is usually not from the vaccine<br />

formulation itself, but from the presence of latex in the equipment used to<br />

hold the vaccine, such as vaccine vial stoppers (bungs) and syringe plungers.<br />

However, very few vaccine bungs contain natural latex. Before administering<br />

the vaccine, consult the product information (PI) of each vaccine to check for the<br />

presence of latex or, where not listed on the PI, contact the vaccine manufacturer<br />

for specific details.<br />

It is important that immunisation service providers assess each individual<br />

for a history of allergies and previous reactions to vaccines prior to giving<br />

any dose of vaccine. Depending on the allergy identified, there often may<br />

not be a contraindication to vaccination. For example, a history of allergy to<br />

antibiotics most commonly relates to β-lactam or related antibiotics and is not a<br />

contraindication to vaccines that contain neomycin, polymyxin B or gentamicin.<br />

Previous reactions to neomycin that only involved the skin are not considered a<br />

risk factor for a severe allergic reaction or anaphylaxis to vaccines manufactured<br />

with neomycin because there are only trace amounts of this antibiotic in the final<br />

product. 3 Similarly, the measles and mumps components of measles-mumpsrubella<br />

(MMR) vaccine contain only a negligible quantity of egg ovalbumin<br />

and do not contraindicate MMR vaccination of persons with egg allergy (even<br />

anaphylaxis) (see ‘Vaccination of persons with a known egg allergy’ below). 4-7<br />

It is important that persons who experience an allergic reaction associated with a<br />

vaccine dose are fully investigated appropriately to ascertain the possible causal<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 131<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS


elationship to vaccination, and determine if, and under what circumstances,<br />

repeat doses of vaccine can be provided. Specialist advice should be sought<br />

where appropriate (see above).<br />

Vaccination of persons with a known egg allergy<br />

Influenza vaccines<br />

A history of anaphylaxis or a severe allergic reaction to eggs has previously been<br />

considered an absolute contraindication to influenza vaccination. However,<br />

there have now been a number of studies indicating that the majority of persons<br />

with egg allergy, including anaphylaxis, can be safely vaccinated with influenza<br />

vaccines that contain less than 1 μg of ovalbumin per dose8-10 (see 4.7 Influenza).<br />

<strong>The</strong> majority of vaccine-associated anaphylaxis cases reported as likely due to<br />

egg allergy occurred following administration of one of the older formulations<br />

of influenza vaccine. 9 Today, due to manufacturing changes, the quantity of<br />

egg ovalbumin present in the majority of influenza vaccines used in Australia<br />

is less than 1 μg of ovalbumin per dose. 8 Note that the amount of residual egg<br />

ovalbumin may vary from year to year due to manufacturing processes, vaccine<br />

batches and country of origin. <strong>The</strong> PI of the vaccine to be given should be<br />

checked for the vaccine’s ovalbumin content prior to vaccine administration. 8,9,11,12<br />

Given that there is still a small risk of anaphylaxis, it is essential that persons<br />

with a history of a severe allergic reaction to eggs are vaccinated in facilities that<br />

have staff who are able to recognise and treat anaphylaxis. 8,9 Allergy testing (e.g.<br />

skin testing) with influenza vaccine prior to administration is not recommended,<br />

as there is poor correlation between test results and vaccine tolerance. 8,9 Detailed<br />

information on influenza vaccination of individuals with an allergy to eggs<br />

can be found in the Australasian Society of Clinical Immunology and Allergy<br />

(ASCIA) guidelines8 (available at www.allergy.org.au/health-professionals/<br />

papers/influenza-vaccination-of-the-egg-allergic-individual).<br />

Other vaccines<br />

Vaccines used in Australia that contain traces of egg ovalbumin, in addition to<br />

most influenza vaccines, are:<br />

• rabies vaccine, Rabipur (see 4.16 Rabies and other lyssaviruses (including<br />

<strong>Australian</strong> bat lyssavirus))<br />

• yellow fever vaccine, Stamaril (see 4.23 Yellow fever)<br />

• Q fever vaccine, Q-Vax (see 4.15 Q fever).<br />

Of these vaccines, yellow fever and Q fever vaccines contain a higher amount of<br />

ovalbumin than is present in the currently available influenza vaccines and are<br />

contraindicated in persons with known severe allergy to eggs. Persons with egg<br />

allergy requiring vaccination with either yellow fever or Q fever vaccines should<br />

seek specialist immunisation advice from state or territory health authorities<br />

132 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


(see Appendix 1 Contact details for <strong>Australian</strong>, state and territory government health<br />

authorities and communicable disease control).<br />

For rabies vaccination, pre- or post-exposure vaccination should be undertaken<br />

using the human diploid cell vaccine (HDCV; Mérieux Inactivated Rabies<br />

Vaccine), and not using the purified chick embryo cell vaccine (PCECV; Rabipur<br />

Inactivated Rabies Virus Vaccine) (see 4.16 Rabies and other lyssaviruses (including<br />

<strong>Australian</strong> bat lyssavirus)).<br />

Although measles and mumps (but not rubella or varicella) vaccine viruses are<br />

grown in chick embryo tissue cultures, it is now recognised that measles- and<br />

mumps-containing vaccines contain negligible amounts of egg ovalbumin<br />

and can be safely administered to persons with a known egg allergy (see 4.9<br />

Measles). 4-7<br />

3.3.2 Vaccination of women who are planning pregnancy,<br />

pregnant or breastfeeding, and preterm infants<br />

Women planning pregnancy<br />

<strong>The</strong> need for vaccination, particularly for hepatitis B, measles, mumps, rubella,<br />

varicella, diphtheria, tetanus and pertussis, should be assessed as part of any<br />

pre-conception health check. Where previous vaccination history or infection is<br />

uncertain, relevant serological testing can be undertaken to ascertain immunity to<br />

hepatitis B, measles, mumps and rubella. Routine serological testing for pertussis<br />

and varicella does not provide a reliable measure of vaccine-induced immunity,<br />

although varicella serology can indicate whether previous natural infection has<br />

occurred (see 4.22 Varicella). Influenza vaccine is recommended for any person<br />

who wishes to be protected against influenza and is recommended for women<br />

planning pregnancy. Those with risk factors for pneumococcal disease, including<br />

smokers and Aboriginal and Torres Strait Islander women, should be assessed<br />

for pneumococcal vaccination. Women who receive live attenuated viral vaccines<br />

should be advised against falling pregnant within 28 days of vaccination.<br />

Refer to the relevant disease-specific chapters in Part 4 for more information<br />

about vaccination requirements for these diseases.<br />

It is also important that women of child-bearing age who present for<br />

immunisation should be questioned regarding the possibility of pregnancy as<br />

part of the routine pre-vaccination screening, to avoid inadvertent administration<br />

of a vaccine(s) not recommended in pregnancy (see 2.1.4 Pre-vaccination<br />

screening).<br />

Pregnant women<br />

Table 3.3.1 summarises the recommendations for vaccine use in pregnancy. More<br />

detailed information is also provided under the ‘Pregnancy and breastfeeding’<br />

sections of each disease-specific chapter in Part 4 of this <strong>Handbook</strong>.<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 133<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS


Seasonal influenza vaccine is the only vaccine routinely recommended for<br />

pregnant women. dTpa vaccine can also be given in pregnancy, as an alternative<br />

to providing it immediately post-partum. Vaccination with dTpa during<br />

pregnancy will provide timely protection against pertussis in both the mother<br />

and her newborn child.<br />

Many other inactivated vaccines are not routinely recommended during<br />

pregnancy on precautionary grounds; however, there is no convincing evidence<br />

that pregnancy should be an absolute contraindication to vaccination with<br />

these vaccines. <strong>The</strong>re is some evidence that fever per se is teratogenic; however,<br />

in clinical studies most inactivated vaccines are not associated with increased<br />

rates of fever in adults (as compared with placebo). 13,14 Recommendations<br />

regarding vaccine use in pregnancy are made where the benefits of protection<br />

from vaccination outweigh the risks. Eliminating the risk of exposure to vaccinepreventable<br />

diseases during pregnancy (e.g. by changing travel plans, avoiding<br />

high-risk behaviours or occupational exposures) is both an alternative and<br />

complementary strategy to vaccination.<br />

Live attenuated viral vaccines are contraindicated in pregnant women because<br />

of the hypothetical risk of harm should vaccine virus replication occur in the<br />

fetus. If a live attenuated viral vaccine is inadvertently given to a pregnant<br />

woman, or if a woman becomes pregnant within 28 days of vaccination, she<br />

should be counselled about the potential for adverse effects, albeit extremely<br />

unlikely, to the fetus (see also 4.18 Rubella and 4.22 Varicella). <strong>The</strong>re is, however,<br />

no indication to consider termination of a pregnancy if a live attenuated vaccine<br />

has been inadvertently given. <strong>The</strong> live attenuated yellow fever vaccine is not<br />

recommended in pregnant women; however, where travel to a yellow fever risk<br />

country is unavoidable, the risks and benefits of yellow fever vaccination, and<br />

other strategies to mitigate the risk of acquiring yellow fever, should be discussed<br />

(see 4.23 Yellow fever).<br />

Inadvertent receipt of a vaccine contraindicated in pregnancy can be reported<br />

to the <strong>The</strong>rapeutic Goods Administration (TGA). For mechanisms for reporting<br />

to the TGA, see 2.3.2 Adverse events following immunisation. Post-marketing<br />

studies of pregnancy outcomes following vaccine administration are important<br />

to understand the safety profile of vaccines in this setting. For this reason some<br />

vaccine manufacturers also operate pregnancy registries, specific for their<br />

products, that will accept reports of vaccines administered during pregnancy; for<br />

example, see information on the varicella vaccine registry in 4.22 Varicella.<br />

134 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


PART 3 VACCINATION FOR SPECIAL RISK GROUPS 135<br />

Table 3.3.1: Recommendations for vaccination in pregnancy (see also disease-specific chapters in Part 4)<br />

Vaccines routinely recommended in pregnancy<br />

Inactivated viral vaccines Recommendation Comments<br />

Influenza vaccine Recommended for all pregnant <strong>The</strong>re is evidence from clinical trial data and observational studies<br />

women at any stage of pregnancy, that there is no increased risk of congenital defects or adverse effects<br />

particularly those who will be in the in the fetuses of women who are vaccinated against influenza in<br />

second or third trimester during the pregnancy. Influenza immunisation protects the mother, as pregnancy<br />

influenza season.<br />

increases her risk of severe influenza, and also protects her newborn<br />

baby in the first few months after birth (see 4.7 Influenza).<br />

Vaccines not routinely recommended in pregnancy<br />

Inactivated bacterial vaccines Recommendation Comments<br />

Diphtheria-, tetanus-, and pertussiscontaining<br />

vaccines (dTpa, dT)<br />

dTpa can be given to pregnant<br />

women in the third trimester as an<br />

alternative to post-partum dTpa (if<br />

a dose of dTpa has not been given in<br />

the previous 5 years).<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS<br />

Vaccination in the third trimester is an acceptable alternative to postpartum<br />

vaccination, for pregnant women who have not been given a<br />

dTpa dose within the previous 5 years. 15 Receipt of dTpa in the third<br />

trimester of pregnancy may be preferred when the risk of the mother<br />

and/or infant acquiring pertussis is high, such as for pregnant women<br />

in close contact with infants. Vaccination during pregnancy has the<br />

advantage of achieving more timely and high pertussis antibody<br />

responses in the mother and infant after birth, as compared with<br />

vaccination given post-partum or prior to conception.<br />

Tetanus- and diphtheria-containing vaccines have been used<br />

extensively in pregnant women, with no increased risk of congenital<br />

abnormalities in fetuses of women who were vaccinated during<br />

pregnancy. 16-18<br />

(See 4.12 Pertussis for more details.) 15<br />

Cholera (oral) vaccine Not routinely recommended <strong>The</strong>re are limited data on the safety of oral cholera vaccine in<br />

pregnancy. 19


136 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Vaccines not routinely recommended in pregnancy<br />

Inactivated bacterial vaccines Recommendation Comments<br />

Haemophilus influenzae type b (Hib)<br />

vaccine<br />

Meningococcal conjugate vaccines<br />

(MenCCV or 4vMenCV)<br />

Meningococcal polysaccharide vaccine<br />

(4vMenPV)<br />

13-valent pneumococcal conjugate<br />

vaccine (13vPCV)<br />

Not routinely recommended<br />

Can be given to pregnant women<br />

at increased risk of Hib disease (e.g.<br />

with asplenia)<br />

Not routinely recommended<br />

Can be given to pregnant women<br />

at increased risk of meningococcal<br />

disease (e.g. with asplenia) or<br />

as post-exposure prophylaxis<br />

in household contacts/cases of<br />

meningococcal serogroup A, C, W 135<br />

or Y<br />

Not routinely recommended<br />

Can be given to pregnant women<br />

at increased risk of meningococcal<br />

disease who have not been<br />

vaccinated with 4vMenPV in the<br />

past 3 years (e.g. with asplenia),<br />

or as post-exposure prophylaxis<br />

in household contacts/cases of<br />

meningococcal serogroup A, W 135<br />

or Y<br />

Limited available data suggest that it is unlikely that use of Hib<br />

vaccine in pregnant women has any deleterious effects on pregnancy<br />

outcomes. 20<br />

<strong>The</strong>re are limited data on the safety of meningococcal conjugate<br />

vaccines in pregnancy. 21 Where clinically indicated, meningococcal<br />

conjugate vaccine can be given to pregnant women. 22<br />

Limited available data suggest that it is unlikely that use of<br />

meningococcal polysaccharide vaccine in pregnant women has<br />

any deleterious effects on pregnancy outcomes. 23,24 Where clinically<br />

indicated, meningococcal polysaccharide vaccine can be given to<br />

pregnant women, although 4vMenCV is preferred. 22<br />

Not routinely recommended No data are available. Vaccination during pregnancy has not been<br />

evaluated, although is unlikely to result in adverse effects.


PART 3 VACCINATION FOR SPECIAL RISK GROUPS 137<br />

Vaccines not routinely recommended in pregnancy<br />

Inactivated bacterial vaccines Recommendation Comments<br />

23-valent pneumococcal polysaccharide<br />

vaccine (23vPPV)<br />

Not routinely recommended<br />

Can be given to pregnant women at<br />

the highest increased risk of invasive<br />

pneumococcal disease (IPD) (e.g.<br />

with asplenia, immunocompromise,<br />

cerebrospinal fluid leak) who have<br />

not received 23vPPV in the past<br />

5 years (and provided they have not<br />

received 2 previous doses)<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS<br />

23vPPV has been administered in pregnancy in the context of clinical<br />

trials 25 with no evidence of adverse effects; however, data are limited.<br />

Where clinically indicated, 23vPPV can be given to pregnant women.<br />

Women of child-bearing age with known risk factors for IPD<br />

(including smokers) should be vaccinated before pregnancy, according<br />

to recommendations (see 4.13 Pneumococcal disease).<br />

Q fever vaccine Not routinely recommended Safe use in pregnancy has not been established.<br />

Typhoid Vi polysaccharide vaccine Not routinely recommended<br />

Can be given to pregnant women<br />

travelling to endemic countries<br />

where water quality and sanitation<br />

is poor<br />

Inactivated viral vaccines Recommendation Comments<br />

Hepatitis A vaccine Not routinely recommended<br />

Can be given to susceptible pregnant<br />

women travelling to areas of<br />

moderate to high endemicity or<br />

those who are at increased risk of<br />

exposure through lifestyle factors,<br />

or where severe outcomes may be<br />

expected (e.g. pre-existing liver<br />

disease)<br />

No data are available. 26 Vaccination during pregnancy has not been<br />

directly evaluated, although is unlikely to result in adverse effects.<br />

Limited data are available.<br />

Hepatitis A vaccine should only be given to pregnant women who are<br />

non-immune and at increased risk for hepatitis A. 27


138 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Vaccines not routinely recommended in pregnancy<br />

Inactivated viral vaccines Recommendation Comments<br />

Hepatitis B vaccine Not routinely recommended<br />

Japanese encephalitis (JE) vaccine<br />

(JEspect)<br />

Can be given to susceptible pregnant<br />

women for whom this vaccine<br />

would otherwise be recommended,<br />

for example, as post-exposure<br />

prophylaxis in a non-immune<br />

pregnant women with a significant<br />

exposure to a HBsAg-positive source<br />

Not routinely recommended<br />

Can be given to pregnant women at<br />

high risk of acquiring JE<br />

Poliomyelitis vaccine (IPV) Not routinely recommended<br />

Can be given to pregnant women at<br />

high risk of poliovirus exposure (e.g.<br />

travel to endemic countries)<br />

Rabies vaccine Can be given to pregnant women for<br />

whom this vaccine would otherwise<br />

be recommended (e.g. post-exposure<br />

prophylaxis).<br />

Limited data are available.<br />

Hepatitis B vaccine should only be given to pregnant women who are<br />

non-immune and at increased risk for hepatitis B. 28<br />

Limited data are available.<br />

JE infection is associated with miscarriage, and women who are at<br />

high risk of JE should be assessed for the need for vaccination. Where<br />

the risk of JE disease is high, pregnant women should be vaccinated<br />

using the inactivated vaccine, JEspect (not Imojev, which is a live<br />

attenuated vaccine). 29<br />

Limited available data suggest that it is unlikely that use of<br />

inactivated poliomyelitis vaccine in pregnant women has any<br />

deleterious effects on pregnancy outcomes. 26<br />

IPV should only be given to pregnant women when clearly indicated.<br />

Limited available data suggest that it is unlikely that the use of rabies<br />

vaccine in pregnant women has any deleterious effects on pregnancy<br />

outcomes. 30-33<br />

Pregnancy is never a contraindication to rabies vaccination in<br />

situations where there is a significant risk of exposure (related to<br />

occupation or travel), or where there has been a potential exposure to<br />

rabies virus, <strong>Australian</strong> bat lyssavirus or another bat lyssavirus. 34,35


PART 3 VACCINATION FOR SPECIAL RISK GROUPS 139<br />

Vaccines not recommended in pregnancy<br />

Inactivated viral vaccines Recommendation Comments<br />

Human papillomavirus (HPV) vaccine Not recommended Although HPV vaccination is not recommended during pregnancy,<br />

evidence from clinical trials and limited data from observational<br />

studies where HPV vaccine was inadvertently administered during<br />

pregnancy, indicate that there is no increased risk of adverse effects on<br />

the fetus. 36<br />

Live attenuated viral vaccines Recommendation Comments<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS<br />

In the event of pregnancy, completion of a 3-dose course of<br />

vaccination should be deferred until after delivery.<br />

Yellow fever vaccine Not recommended Pregnant women should be advised against going to the rural areas<br />

of yellow fever endemic areas. However, where travel to an at-risk<br />

country is unavoidable, such women should be vaccinated. 37,38 Yellow<br />

fever vaccine has been given to a large number of pregnant women<br />

with no adverse outcomes. 39<br />

Vaccines contraindicated in pregnancy<br />

Live attenuated bacterial vaccines Recommendation Comments<br />

BCG vaccine Contraindicated <strong>The</strong>re is only a hypothetical risk. BCG vaccine has not been shown to<br />

cause fetal damage. 40<br />

Oral typhoid vaccine Contraindicated <strong>The</strong>re are limited data available (animal studies), suggesting no<br />

increased occurrence of fetal damage with oral live attenuated<br />

vaccine. 41 Inactivated typhoid Vi polysaccharide vaccine is preferred<br />

(see above).<br />

Live attenuated viral vaccines Recommendation Comments<br />

Japanese encephalitis (JE) vaccine<br />

(Imojev)<br />

Contraindicated <strong>The</strong>re is only a hypothetical risk. <strong>The</strong>re are currently no data available<br />

regarding the use of this vaccine in pregnant or breastfeeding women.<br />

Women of child-bearing age should avoid pregnancy for 28 days after<br />

vaccination.


140 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Vaccines contraindicated in pregnancy<br />

Live attenuated viral vaccines Recommendation Comments<br />

Measles-mumps-rubella (MMR) Contraindicated <strong>The</strong>re is only a hypothetical risk. Despite concerns that live attenuated<br />

vaccine<br />

rubella vaccine virus might cause congenital abnormalities, rubella<br />

or<br />

vaccine (either monovalent or as MMR) has been given to pregnant<br />

Measles-mumps-rubella-varicella<br />

women (usually inadvertently) without harm to the fetus.<br />

(MMRV) vaccine<br />

42,43 Even<br />

though rubella vaccine virus can infect the fetus, even for vaccine<br />

given in early pregnancy, there is no evidence that it causes congenital<br />

rubella syndrome in infants born to susceptible mothers. 44 Receipt<br />

of rubella vaccination during pregnancy is not an indication for<br />

termination. 43<br />

Women of child-bearing age should avoid pregnancy for 28 days after<br />

vaccination.<br />

It is recommended practice to test all pregnant women for immunity<br />

to rubella, and to vaccinate susceptible women as soon as possible<br />

after delivery and check their serological status post vaccination.<br />

Rotavirus vaccine Contraindicated Rotavirus vaccines are not registered or recommended for use in<br />

adolescents or adults.<br />

Varicella vaccine Contraindicated <strong>The</strong>re is only a hypothetical risk. Congenital varicella syndrome has<br />

not been identified in women who have been inadvertently vaccinated<br />

with varicella vaccine in early pregnancy. 45<br />

Women of child-bearing age should avoid pregnancy for 28 days after<br />

vaccination.<br />

Zoster vaccine Contraindicated <strong>The</strong>re is only a hypothetical risk. Women of child-bearing age are<br />

unlikely to be eligible for vaccination, as zoster vaccine is registered<br />

for use in persons ≥50 years of age. If women of child-bearing age<br />

have inadvertently been vaccinated, they should avoid pregnancy for<br />

28 days after vaccination.


PART 3 VACCINATION FOR SPECIAL RISK GROUPS 141<br />

Immunoglobulins for use as pre- or post-exposure prophylaxis<br />

Pooled or hyperimmune<br />

immunoglobulins<br />

Not routinely recommended<br />

Can be used post exposure in<br />

susceptible pregnant women<br />

exposed to: measles, hepatitis A,<br />

hepatitis B, rabies, <strong>Australian</strong> bat<br />

lyssavirus, or varicella viruses, or<br />

tetanus<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS<br />

Limited data are available.<br />

<strong>The</strong>re is no known risk to the fetus from passive immunisation of<br />

pregnant women with immunoglobulins.<br />

For more details, see Part 5 Passive immunisation and relevant diseasespecific<br />

chapters in Part 4.


Contact between pregnant women and persons who have recently received live vaccines<br />

Household contacts of pregnant women should be age-appropriately vaccinated.<br />

It is safe to administer measles-, mumps-, rubella- and varicella-containing<br />

vaccines, zoster vaccine and rotavirus vaccine to the contacts of pregnant<br />

women. <strong>The</strong>re is no risk of transmission of measles, mumps or rubella vaccine<br />

viruses from vaccinated household contacts. <strong>The</strong>re is an almost negligible risk<br />

of transmission of varicella-zoster vaccine virus (from persons vaccinated with<br />

varicella or zoster vaccines); however, vaccine recipients with a varicella-like<br />

rash should be advised to cover the rash if in contact with a pregnant woman.<br />

Although there is a very small possibility of transmission of the rotavirus vaccine<br />

viruses to pregnant contacts, the benefit of immunising infants to protect against<br />

rotavirus disease and, in turn, reduce the risk of rotavirus in household contacts,<br />

far outweighs any theoretical risk (see 4.17 Rotavirus).<br />

Use of immunosuppressive therapy during pregnancy<br />

Certain immunosuppressive medications given for management of a medical<br />

condition in a woman during pregnancy (e.g. biological disease modifying<br />

anti-rheumatic drugs [bDMARDs]) may cross the placenta and be detectable<br />

in the infant, particularly if given during the third trimester. 46-48 In this setting,<br />

administration of live attenuated vaccines in the first few months of the infant’s<br />

life, particularly BCG vaccine, is not recommended. 49 (See also 4.20 Tuberculosis.)<br />

This is because of the risk that the infant’s immune response to vaccination may<br />

be reduced and potentially associated with increased vaccine virus/bacteria<br />

replication and related adverse effects. Although no specific time intervals<br />

are indicated, withholding BCG vaccine until the infant is 6 months of age is<br />

prudent. 50 Although there is some theoretical concern that a risk also applies<br />

to the administration of rotavirus vaccines, there are currently no data to<br />

substantiate this.<br />

Inactivated vaccines should be administered to these infants according to the<br />

recommended schedule. However, immune responses may be suboptimal.<br />

Additional inactivated vaccine doses may be required; expert advice should be<br />

sought regarding this.<br />

Breastfeeding and vaccination<br />

Vaccination is rarely contraindicated in breastfeeding women. <strong>The</strong> rubella<br />

vaccine virus may be secreted in human breast milk and there has been<br />

documented transmission to breastfed infants. However, where infection has<br />

occurred in an infant, the symptoms have been absent or mild. 51-53 Infants born<br />

to mothers who are hepatitis B surface antigen (HBsAg)-positive can also be<br />

breastfed, provided the infant is appropriately immunised at birth. Although<br />

studies have indicated the presence of hepatitis B virus (HBV) in the breast<br />

milk of mothers with HBV infection, breastfeeding poses no additional risk of<br />

virus transmission, compared with formula feeding, in vaccinated infants. 54<br />

Administration of yellow fever vaccine to breastfeeding women should be<br />

142 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


avoided, except in situations where the risk of acquiring yellow fever is high,<br />

and/or travel cannot be avoided or postponed. 55,56 While extremely rare, there<br />

have been several case reports of probable transmission of the yellow fever<br />

vaccine virus via breast milk. 55,56 For most vaccines, the immune response to<br />

vaccination of infants in relationship to breastfeeding has been studied and taken<br />

into account. In general, breastfeeding does not adversely affect immunisation,<br />

and breastfeeding is not a contraindication to the administration of any vaccines<br />

recommended in infants.<br />

Preterm infants<br />

Preterm (premature) infants are defined as those born at


doses after age 5 years. However, all children and adults who have chronic lung<br />

disease, or certain other chronic medical conditions, whether related to preterm<br />

birth or not, should also receive additional pneumococcal vaccine doses up to and<br />

beyond the age of 5 years (see 4.13 Pneumococcal disease).<br />

Hepatitis B vaccine<br />

Low-birth-weight preterm newborn infants do not respond as well to hepatitis<br />

B-containing vaccines as full-term infants. 69,74,75 Thus, for low-birth-weight<br />

infants (


3.3.3 Vaccination of immunocompromised persons<br />

A person can be immunocompromised due to disease and/or medical<br />

treatment. Vaccination of immunocompromised persons presents numerous<br />

challenges. <strong>The</strong> immune protection attained from previous immunisation<br />

may be diminished; the response to vaccines administered in the setting of<br />

immunocompromise may be reduced, with additional booster vaccine doses<br />

required; the risk of vaccine-preventable diseases and/or their complications<br />

may be increased; and the risk of adverse events from live vaccines may be<br />

increased. Degrees of immunocompromise vary from insignificant to profound,<br />

and this, together with the risk of acquiring vaccine-preventable disease, should<br />

be taken into account when considering a vaccination schedule.<br />

When considering vaccination of persons on immunosuppressive therapy, it is<br />

particularly important to consider a number of factors, including the biologic<br />

target of the medication being used (mechanism and duration of effect on the<br />

immune system) as well as the consequence of using combination therapies<br />

(e.g. prednisolone and methotrexate), which can contribute to the nature,<br />

extent and length of immunocompromise. It is also important to know the<br />

anticipated duration of immunocompromise, whether due to therapy or the<br />

underlying disease (see also ‘Immunocompromise associated with corticosteroid<br />

administration’ below). In some instances, additional booster doses of vaccines<br />

may be required to optimise protection in immunocompromised persons<br />

(e.g. pneumococcal vaccines at diagnosis of haematological malignancy).<br />

To determine the need for booster doses, it may be useful to measure postvaccination<br />

antibody titres in selected groups in some circumstances, such as<br />

for adults or children who have received haematopoietic stem cell transplants<br />

(see ‘Haematopoietic stem cell transplant recipients’ below). Reliable serological<br />

testing is not readily available and/or validated to measure vaccine-induced<br />

immunity for all vaccines, and, in addition, results should be interpreted using<br />

standardised serological correlates. (See also 2.1.5 Catch-up, ‘Use of serological<br />

testing to guide catch-up vaccination’.) Expert advice should be sought if<br />

required.<br />

Many vaccine-preventable diseases are associated with an increased risk of<br />

morbidity and mortality in immunocompromised persons. Two important<br />

examples are influenza and invasive pneumococcal disease (IPD). Annual<br />

influenza vaccination should be given to all immunocompromised persons<br />

≥6 months of age (see 4.7 Influenza). Immunocompromised persons may<br />

also require additional doses of pneumococcal vaccines; the timing, number<br />

of doses and type of vaccine(s) vary depending on age and the underlying<br />

risk for IPD (see 4.13 Pneumococcal disease). <strong>The</strong>se, and other specific vaccine<br />

recommendations, are discussed in more detail below.<br />

All immunocompromised persons, irrespective of age, who receive influenza<br />

vaccine for the first time are recommended to receive 2 vaccine doses, at least<br />

4 weeks apart, and 1 dose annually thereafter (see 4.7 Influenza). Where it is<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 145<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS


known that a new influenza vaccine strain is circulating in the community to<br />

which cross-protective immunity in the population is low (such as in the setting<br />

of an influenza pandemic), it may be appropriate that immunocompromised<br />

persons receive 2 doses of inactivated influenza vaccine, a minimum of 4 weeks<br />

apart, to achieve an optimal immune response. For example, in the 2009–2010<br />

H1N1 global influenza pandemic it was shown that seroconversion to influenza<br />

vaccination in immunocompromised adolescents and adults was improved<br />

following receipt of 2 vaccine doses. 77 Further information and annual influenza<br />

vaccine recommendations are available on the Immunise Australia website<br />

(www.immunise.health.gov.au).<br />

<strong>The</strong> recommendations in this section for the use of vaccines in<br />

immunocompromised persons have been divided where applicable into<br />

paediatric (0–18 years) and adult (≥19 years) recommendations. This distinction<br />

has been made on the basis of scientific evidence, where available, and to assist in<br />

vaccine delivery in paediatric and adult special risk settings.<br />

Immunocompromise associated with corticosteroid administration<br />

<strong>The</strong> dose and duration of therapy with corticosteroids determines the impact<br />

on the immune system. In adults, daily doses of oral corticosteroids in excess of<br />

60 mg of prednisolone (or equivalent) for more than 1 week are associated with<br />

significant immunocompromise. In children, doses in excess of either<br />

2 mg/kg per day for more than 1 week or 1 mg/kg per day for more than<br />

4 weeks are associated with significant immunocompromise. Live attenuated<br />

vaccines are generally contraindicated in such persons (see also below). In<br />

addition, for both children and adults, even lower doses may be associated with<br />

some impairment of the immune response. 78 It is also important, once treatment<br />

with corticosteroids is ceased, to assess whether the person has other underlying<br />

immunocompromising disease or is receiving other immunosuppressive therapy<br />

that may influence decisions about whether vaccines, particularly live vaccines,<br />

can be given.<br />

For adults treated with systemic corticosteroids in excess of 60 mg per day for<br />

more than 1 week, live attenuated viral vaccines (such as MMR, MMRV, zoster,<br />

varicella and yellow fever vaccines) should be postponed until at least 1 month<br />

after treatment has stopped.<br />

Children receiving >2 mg/kg per day or ≥20 mg per day in total of prednisolone<br />

(or equivalent) for more than 1 week should not receive live attenuated vaccines<br />

until after corticosteroid therapy has been discontinued for at least 1 month.<br />

Children on daily doses of ≤2 mg/kg per day of systemic corticosteroids for<br />

less than 1 week, and those on lower doses of 1 mg/kg per day or alternateday<br />

regimens for periods of up to 4 weeks, may be given live attenuated viral<br />

vaccines. Some experts suggest withholding lower doses of steroids 2 to 3 weeks<br />

prior to vaccination with live viral vaccines if this is possible. 79,80<br />

146 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Use of live viral or live bacterial vaccines in immunocompromised persons<br />

<strong>The</strong>re is a risk that the administration of live vaccines to immunocompromised<br />

persons may result in adverse events or vaccine-related disease due to<br />

unchecked infection (replication) of the vaccine virus or bacteria. This is<br />

particularly so for measles-, mumps-, rubella-81,82 and VZV-containing<br />

(varicella and zoster) vaccines 83 and for bacille Calmette-Guérin (BCG)<br />

vaccine. 49,84 However, the risk of disease varies by vaccine and by individual.<br />

Caution is required for vaccination in the setting of immunocompromise,<br />

and in significantly immunocompromised persons most live vaccines are<br />

contraindicated.<br />

<strong>The</strong> following is a list of current recommendations for use of live vaccines in<br />

immunocompromised persons.<br />

• Tuberculosis vaccine (BCG) is always contraindicated.<br />

• Live vaccines, such as MMR- and VZV-containing (varicella and zoster)<br />

vaccines, should not be given to persons with severe immunocompromise.<br />

Severely immunocompromised persons include those who have active<br />

leukaemia or lymphoma, generalised malignancy, aplastic anaemia,<br />

graft-versus-host disease or congenital immunodeficiency. Others in<br />

this category include persons who have received recent chemotherapy,<br />

persons who have had solid organ or bone marrow transplants (within<br />

2 years of transplantation) or transplant recipients who are still taking<br />

immunosuppressive drugs, or others on highly immunosuppressive therapy,<br />

including high-dose corticosteroids (see above). Dependent on their age,<br />

persons infected with human immunodeficiency virus (HIV) with CD4 + cell<br />

counts of


• Yellow fever vaccine is generally contraindicated in immunocompromised<br />

travellers going to yellow fever endemic countries. <strong>The</strong> vaccine can, however,<br />

be considered on a case-by-case basis, including in persons with HIV (see<br />

4.23 Yellow fever).<br />

If there is uncertainty around the level of immunocompromise and when vaccine<br />

administration may be safe, this should be discussed with the treating physician<br />

and expert advice should be sought (see also ‘Immunocompromise associated<br />

with corticosteroid administration’ above).<br />

Household contacts of immunocompromised persons<br />

To best protect immunocompromised persons, whether adults or children, their<br />

household and other close contacts should be fully vaccinated according to<br />

current recommendations. Annual influenza vaccination is recommended for<br />

all household contacts (≥6 months of age) of immunocompromised persons.<br />

Assessment of the need for household contacts of immunocompromised persons<br />

to receive pertussis-containing and/or varicella vaccines is also very important<br />

(see 4.12 Pertussis and 4.22 Varicella). 85-87<br />

<strong>The</strong> use of live attenuated viral vaccines in contacts of immunocompromised<br />

persons (MMR, MMRV, varicella and rotavirus vaccines, where indicated) is<br />

safe, and strongly recommended to reduce the likelihood of contacts infecting<br />

the immunocompromised person. Persons ≥50 years of age who are household<br />

contacts of an immunocompromised person are also recommended to receive<br />

zoster vaccine. Although there is no risk of transmission of the MMR vaccine<br />

viruses, and an almost negligible risk of transmission of varicella-zoster vaccine<br />

virus (from varicella or zoster vaccine), there is a small risk of transmission of the<br />

rotavirus vaccine virus. Hand washing and careful disposal of soiled nappies is<br />

recommended to minimise transmission. Immunocompromised persons should<br />

avoid contact with persons with varicella and herpes zoster, where possible. (See<br />

also 4.9 Measles, 4.17 Rotavirus, 4.22 Varicella and 4.24 Zoster).<br />

Oncology patients<br />

Paediatric and adult patients undergoing cancer chemotherapy who have not completed a<br />

primary vaccination schedule before diagnosis<br />

Live vaccines, including BCG, MMR, zoster and varicella vaccines, are<br />

contraindicated in cancer patients receiving immunosuppressive therapy and/or<br />

who have poorly controlled malignant disease. <strong>The</strong>se vaccines are recommended<br />

to be administered to seronegative persons at least 3 months after completion<br />

of chemotherapy, provided the underlying malignancy is in remission. 88<br />

Administration of live attenuated viral vaccines (MMR-containing or varicellacontaining<br />

vaccines) should be deferred if blood products or immunoglobulins<br />

have been recently administered (see Table 3.3.6 Recommended intervals between<br />

either immunoglobulins or blood products and MMR, MMRV or varicella vaccination).<br />

148 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Influenza vaccination is recommended annually in all cancer patients aged<br />

≥6 months. All immunocompromised persons, irrespective of age, who receive<br />

influenza vaccine for the first time are recommended to receive 2 vaccine doses,<br />

at least 4 weeks apart, and 1 dose annually thereafter.<br />

Persons receiving chemotherapy may receive inactivated vaccines (e.g. 13vPCV,<br />

hepatitis B) according to a routine or catch-up vaccination schedule. <strong>The</strong> immune<br />

response may be suboptimal, but the vaccines are safe to administer.<br />

Vaccines should not be administered during times of severe neutropenia<br />

(absolute neutrophil count


• varicella vaccine: persons who are seronegative to varicella-zoster virus<br />

(VZV) should receive a 2-dose schedule of varicella vaccine, at least 6 months<br />

after chemotherapy has ceased (see 4.22 Varicella).<br />

Measles and rubella antibody status should be checked 6 to 8 weeks after<br />

vaccination with MMR or MMRV vaccine. Persons who have not seroconverted<br />

should receive a further dose.<br />

Administration of live attenuated viral vaccines (MMR-containing or varicellacontaining<br />

vaccines) should be deferred if blood products or immunoglobulins<br />

have been recently administered (see Table 3.3.6 Recommended intervals between<br />

either immunoglobulins or blood products and MMR, MMRV or varicella vaccination).<br />

Solid organ transplant recipients<br />

For solid organ transplant (SOT) recipients, depending on the transplanted<br />

organ, and to prevent rejection, variable doses of immunosuppressive agents<br />

are required and may influence the effectiveness of vaccines. Where possible,<br />

children undergoing solid organ transplantation should be vaccinated well<br />

before transplantation. Inactivated vaccines can be administered safely after<br />

transplantation, but are usually administered from 6 months after transplantation<br />

to maximise the immune response. 94,95 Live vaccines are contraindicated in<br />

most post-transplantation protocols due to concerns of disseminated infection,<br />

although data in this population are limited. 95-97 Recommended vaccinations for<br />

child and adult SOT recipients are given in Table 3.3.2.<br />

150 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


PART 3 VACCINATION FOR SPECIAL RISK GROUPS 151<br />

Table 3.3.2: Recommendations for vaccinations for solid organ transplant (SOT) recipients 96,98<br />

Vaccine Vaccines recommended before<br />

transplantation<br />

Child<br />

(0–18 years)<br />

Adult<br />

(≥19 years)<br />

Streptococcus pneumoniae (pneumococcal disease)<br />

13-valent<br />

pneumococcal<br />

conjugate vaccine<br />

(13vPCV)<br />

23-valent<br />

pneumococcal<br />

polysaccharide<br />

vaccine<br />

(23vPPV)<br />

Yes (aged<br />

≥6 weeks)<br />

Yes (≥8 weeks<br />

after 13vPCV)<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS<br />

Vaccines recommended after<br />

transplantation, if not given<br />

beforehand<br />

Child<br />

(0–18 years)<br />

Yes Yes (aged<br />

≥6 weeks)<br />

Yes (≥8 weeks<br />

after 13vPCV)<br />

Yes (≥8 weeks<br />

after 13vPCV)<br />

Adult<br />

(≥19 years)<br />

Comment<br />

Yes Recommendations depend on age. See 4.13<br />

Pneumococcal disease and Table 2.1.11 Catch-up schedule<br />

for 13vPCV (Prevenar 13) and 23vPPV (Pneumovax 23)<br />

in children with a medical condition(s) associated with an<br />

increased risk of IPD, presenting at age


152 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Vaccine Vaccines recommended before<br />

transplantation<br />

Child<br />

(0–18 years)<br />

Adult<br />

(≥19 years)<br />

Vaccines recommended after<br />

transplantation, if not given<br />

beforehand<br />

Child<br />

(0–18 years)<br />

Adult<br />

(≥19 years)<br />

Comment<br />

Influenza<br />

Influenza vaccine Annual vaccination starting before transplantation for those ≥6 months of age. Two doses of influenza vaccine at least 4 weeks<br />

apart are recommended for all SOT recipients receiving influenza vaccine for the first time. Influenza vaccine should be given<br />

annually thereafter.<br />

Poliomyelitis<br />

IPV Yes Yes (see<br />

comments)<br />

Hepatitis B<br />

Hepatitis B vaccine Yes Yes, depending<br />

on serological<br />

status<br />

Yes Yes (see<br />

comments)<br />

Yes Yes, depending on<br />

serological status<br />

Hepatitis A<br />

Hepatitis A vaccine* Yes, if<br />

Yes, if<br />

Yes, if<br />

Yes, if<br />

seronegative (see seronegative (see seronegative (see seronegative (see<br />

comments) comments) comments) comments)<br />

Adults who have received a routine course of polio<br />

vaccination in childhood are recommended to receive<br />

a booster every 10 years if they plan to travel to a<br />

polio endemic area or have an occupational risk of<br />

polio exposure (e.g. laboratory workers).<br />

Recommended for all seronegative SOT candidates.<br />

Immunogenicity is likely to be improved when<br />

vaccination is administered before transplantation.<br />

Accelerated schedules can be used (see Table 4.5.2<br />

Accelerated hepatitis B vaccination schedules (for persons<br />

with imminent risk of exposure)).<br />

Recommended for all liver SOT recipients, or<br />

transplant candidates or recipients with chronic liver<br />

disease, or those chronically infected with either<br />

hepatitis B or hepatitis C.


PART 3 VACCINATION FOR SPECIAL RISK GROUPS 153<br />

Vaccine Vaccines recommended before<br />

transplantation<br />

Child Adult<br />

(0–18 years) (≥19 years)<br />

Neisseria meningitidis (meningococcal disease)<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS<br />

Vaccines recommended after<br />

transplantation, if not given<br />

beforehand<br />

Child Adult<br />

(0–18 years) (≥19 years)<br />

Comment<br />

Meningococcal C Yes Not indicated Yes Not indicated If the 1st MenCCV dose is given before 12 months of<br />

conjugate vaccine age, a 2nd dose should be given at least 8 weeks later.<br />

(MenCCV) Give 4vMenCV, if clinically indicated (see below),<br />

once child is ≥12 months of age, and at least 8 weeks<br />

has elapsed since receipt of last MenCCV dose.<br />

Quadrivalent Yes, if ≥9 months Yes, if defined Yes, if ≥9 months Yes, if defined 4vMenCV as a 2-dose schedule is recommended as a<br />

meningococcal of age with risk factors (see of age with risk factors (see primary course of vaccination for those (≥9 months of<br />

conjugate vaccine defined risk comments) defined risk comments) age) with complement component deficiencies (e.g.<br />

(4vMenCV)* factors (see<br />

comments)<br />

factors (see<br />

comments)<br />

Human papillomavirus<br />

HPV vaccine Yes Yes Yes, if no<br />

history of prior<br />

immunisation<br />

Measles, mumps and rubella<br />

MMR vaccine Yes Yes, unless<br />

2 previous<br />

documented<br />

doses<br />

Yes, if no<br />

history of prior<br />

immunisation<br />

C5-C9, properdin, Factor D, Factor H), functional<br />

hyposplenism or anatomical asplenia (see Table<br />

3.3.5 Recommendations for vaccination in persons with<br />

functional or anatomical asplenia). Boosters of 4vMenCV<br />

should be given every 5 years.<br />

3-dose schedule of 4vHPV is recommended for those<br />

aged >9 years. <strong>The</strong> routine schedule is 1st dose on day<br />

0 (day of vaccination), 2 months, and 6 months (after<br />

1st dose). Recommended in both females and males.<br />

For more detail, see 4.6 Human papillomavirus.<br />

Contraindicated Contraindicated <strong>The</strong> primary schedule should be completed before<br />

transplantation provided the transplant candidate is<br />

taking no immunosuppressive therapy and has no<br />

underlying cellular immunodeficiency.


154 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Vaccine Vaccines recommended before<br />

transplantation<br />

Child<br />

(0–18 years)<br />

Varicella<br />

Varicella vaccine Yes, if nonimmune<br />

(see<br />

comments)<br />

Adult<br />

(≥19 years)<br />

Yes, if nonimmune<br />

(see<br />

comments)<br />

Vaccines recommended after<br />

transplantation, if not given<br />

beforehand<br />

Child<br />

(0–18 years)<br />

Adult<br />

(≥19 years)<br />

Comment<br />

Contraindicated Contraindicated Confirm immunity with reliable history of varicella<br />

disease and confident clinical diagnosis or serological<br />

testing.<br />

<strong>The</strong> primary vaccination schedule should be<br />

completed before transplantation, provided the<br />

transplant candidate is taking no immunosuppressive<br />

therapy and has no underlying cellular<br />

immunodeficiency.<br />

* Any transplant recipient who anticipates travelling may require additional vaccination, such as for hepatitis A<br />

and meningococcal disease (see also 3.2 Vaccination for international travel).


Haematopoietic stem cell transplant recipients 99,100<br />

Haematopoietic stem cells are sourced from peripheral blood, bone marrow<br />

or umbilical cord blood. Protective immunity to vaccine-preventable diseases<br />

is partially or completely lost following either allogeneic or autologous stem<br />

cell transplantation. Immunocompromise following allogeneic transplantation<br />

is caused by a combination of the preparative chemotherapy given before<br />

transplantation, graft-versus-host disease (GVHD), and immunosuppressive<br />

therapy following transplantation. Persisting immunocompromise is common,<br />

particularly in persons with chronic GVHD. Immunity is also impaired in<br />

autologous HSCT recipients due to high-dose chemotherapy and radiotherapy,<br />

but GVHD is not a concern as donor stem cells are derived from the transplant<br />

recipient. In most cases, autologous HSCT recipients will recover their immunity<br />

more quickly than allogeneic transplant recipients.<br />

Separate vaccination schedules for autologous or allogeneic HSCT recipients<br />

have not been supported in published guidelines because of limited data. For<br />

practical purposes, the same schedule is recommended for these two groups,<br />

regardless of donor source (peripheral blood, bone marrow or umbilical cord),<br />

preparative chemotherapy (ablative or reduced intensity), or transplant type<br />

(allogeneic or autologous). 101,102<br />

HSCT recipients with ongoing GVHD or remaining on immunosuppressive<br />

therapy should not be given live vaccines. Chronic GVHD (cGVHD) is associated<br />

with functional hyposplenism and therefore increases susceptibility to infections<br />

with encapsulated organisms, especially Streptococcus pneumoniae. For persons<br />

with cGVHD who remain on active immunosuppression, antibiotic prophylaxis<br />

is recommended. 99<br />

<strong>The</strong> immune response to vaccinations is usually poor during the first 6 months<br />

after HSCT. Donor immunisation with hepatitis B, tetanus, Hib and<br />

pneumococcal conjugate vaccines before stem cell harvesting has been shown to<br />

elicit improved early antibody responses in HSCT recipients vaccinated in the<br />

post-transplantation period. 103-106 However, practical and ethical considerations<br />

currently limit the use of donor immunisation.<br />

Routine serological testing for several infectious agents and antibody levels<br />

conferring protective immunity are poorly defined. For those vaccines that<br />

are recommended for all HSCT recipients (tetanus, diphtheria, poliomyelitis,<br />

influenza, pneumococcal, Hib), pre-vaccination testing is not recommended as<br />

the response to a primary course of these vaccines is generally adequate. <strong>The</strong><br />

serological response to pneumococcal vaccine is less predictable. Pneumococcal<br />

serology is only available in a few specialised laboratories and is not routinely<br />

recommended. Serology before and approximately 4 to 6 weeks after vaccination<br />

with the final dose of a hepatitis B vaccine course, and after MMR vaccine, is<br />

recommended as antibody levels will determine the need for revaccination. 102<br />

Post-vaccination varicella serology using commercial assays is very insensitive<br />

for vaccine-induced immunity (as compared with natural infection) and is not<br />

recommended (see 4.22 Varicella).<br />

A recommended schedule of vaccination is outlined in Table 3.3.3.<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 155<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS


Table 3.3.3: Recommendations for revaccination following haematopoietic<br />

stem cell transplant (HSCT) in children and adults, irrespective of<br />

previous immunisation history 99,100,107-111<br />

Vaccine Months after HSCT Comments<br />

6 8 12 24<br />

Streptococcus pneumoniae (pneumococcal disease)<br />

13-valent<br />

pneumococcal<br />

conjugate<br />

vaccine<br />

(13vPCV)<br />

23-valent<br />

pneumococcal<br />

polysaccharide<br />

vaccine<br />

(23vPPV)<br />

Yes Yes Yes Not needed See 4.13 Pneumococcal disease<br />

No No No Yes (after<br />

13vPCV)<br />

156 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

See 4.13 Pneumococcal disease<br />

Haemophilus influenzae type b<br />

Hib Yes Yes Yes Not needed<br />

Diphtheria, tetanus, pertussis<br />

DTPa-containing Yes Yes Yes Not needed For recipients


Vaccine Months after HSCT Comments<br />

6 8 12 24<br />

Neisseria meningitidis (meningococcal disease)<br />

Meningococcal<br />

C conjugate<br />

vaccine<br />

(MenCCV)<br />

(for those<br />

12 months post<br />

transplantation when<br />

a greater level of<br />

immune reconstitution<br />

has been achieved.<br />

Measles, mumps and rubella<br />

MMR vaccine No No No Yes, 1 or<br />

2 doses<br />

separated by<br />

a minimum<br />

interval of<br />

4 weeks (see<br />

comments)<br />

Varicella<br />

Varicella vaccine No No No Yes, 2 doses<br />

separated by<br />

a minimum<br />

interval of<br />

4 weeks (see<br />

comments)<br />

Not needed Two doses of 4vMenCV are<br />

recommended for persons<br />

≥12 months of age (see 4.10<br />

Meningococcal disease).<br />

Individual recommendations for<br />

HPV vaccination in those >9 years<br />

of age should be determined by an<br />

individual risk assessment (see 4.6<br />

Human papillomavirus).<br />

Give only if the person is off<br />

immunosuppressive therapy, with<br />

no cGVHD and with reconstituted<br />

cell-mediated immunity. Check<br />

serology 4 weeks after 1st vaccine<br />

dose. If there is no seroconversion,<br />

repeat the dose.<br />

Give to a seronegative recipient<br />

only if the person is off<br />

immunosuppressive therapy, with<br />

no cGVHD and with reconstituted<br />

cell-mediated immunity.<br />

* Any transplant recipient who anticipates travelling may require additional vaccination, such as<br />

for meningococcal and hepatitis A disease (see also 3.2 Vaccination for international travel).<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 157<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS


HIV-infected persons 112<br />

Vaccination schedules for HIV-infected persons should be determined by the<br />

person’s age, degree of immunocompromise (CD4 + count) and the risk of<br />

infection (see Table 3.3.4 below). Children with perinatally acquired HIV differ<br />

substantially from adults, as immunisation and first exposure to vaccine antigens<br />

occurs after HIV infection, whereas in adults, most vaccines are inducing a<br />

secondary ‘boosted’ immune response. HIV-infected persons of any age whose<br />

disease is well controlled on combination antiretroviral therapy (undetected or<br />

low viral load with good preservation of CD4 + lymphocyte count) are likely to<br />

respond satisfactorily to vaccines.<br />

Table 3.3.4: Categories of immunocompromise in HIV-infected persons, based<br />

on age-specific CD4 + counts and percentage of total lymphocytes 113<br />


depending on the number of vaccines received previously and evidence for<br />

seroconversion. Administration of MMR vaccine does not have a significant<br />

effect on the CD4 + count or viral load of HIV-infected adults. 121 Measles may<br />

cause severe disease in HIV-infected children, particularly those with a CD4 +<br />

count of 200 per µL, who are at a recognised risk of exposure; however, this<br />

should be discussed with the person’s treating clinician. 128 (See also 4.23<br />

Yellow fever and 3.2 Vaccination for international travel.)<br />

• BCG vaccine should not be given to HIV-infected children or adults because<br />

of the risk of disseminated BCG infection. 129,130 (See also 4.20 Tuberculosis.)<br />

• Oral live attenuated typhoid vaccines should be avoided in HIV-infected<br />

persons. Parenteral Vi polysaccharide typhoid vaccine should be used instead<br />

(see 4.21 Typhoid).<br />

Inactivated (non-live) vaccines<br />

• Diphtheria-tetanus-pertussis (DTPa/dTpa), Hib and IPV vaccines can be<br />

given according to routine recommendations112,131 (see relevant diseasespecific<br />

chapters in Part 4).<br />

• <strong>The</strong> 4vHPV vaccine can be given to children (≥9 years of age) and adults with<br />

HIV. It was safe and immunogenic in a small study of HIV1-infected men. 132<br />

HIV-infected persons should receive the routine course of 3 doses of 4vHPV<br />

vaccine at times 0, 2 and 6 months. Vaccination is recommended for persons<br />

in the age ranges for which the vaccine is registered (females aged 9–45 years<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 159<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS


and males 9–26 years); use of HPV vaccine in males up to the age of 45 years<br />

is unlikely to be associated with immunogenicity or adverse events that differ<br />

from those observed in females. However, the benefit of HPV vaccination<br />

is optimal when delivered to children or young adolescents prior to sexual<br />

debut (see 4.6 Human papillomavirus).<br />

• Pneumococcal disease, both respiratory and invasive (IPD), is a frequent<br />

cause of morbidity in HIV-infected children and adults (see List 4.13.1 in<br />

4.13 Pneumococcal disease). 133 Children should be vaccinated initially with<br />

pneumococcal conjugate vaccine (13vPCV); the number of doses depends<br />

on age at diagnosis and vaccination history (see Table 2.1.11 Catch-up<br />

schedule for 13vPCV (Prevenar 13) and 23vPPV (Pneumovax 23) in children with<br />

a medical condition(s) associated with an increased risk of IPD, presenting at age<br />

5 years and adults, a single dose of 13vPCV<br />

is recommended, followed by 23vPPV; repeat doses of 23vPPV are also<br />

indicated. See 4.13 Pneumococcal disease for details.<br />

• Annual influenza vaccination is recommended in all HIV-infected<br />

adults and children (≥6 months of age). In HIV-infected persons who are<br />

immunocompromised and children


Persons with functional or anatomical asplenia<br />

Persons with an absent or dysfunctional spleen are at a life-long increased risk<br />

of fulminant bacterial infection, most notably invasive pneumococcal disease<br />

(IPD). 89,141 Pneumococcal, meningococcal, Hib and influenza vaccination are<br />

particularly recommended for all persons with asplenia, whether functional<br />

or anatomical (such as splenectomy). Other vaccinations should be up to<br />

date. Vaccines should be provided according to the person’s age and previous<br />

immunisation history, and immunisation status should be reviewed regularly. 142<br />

Specific vaccine recommendations for persons with asplenia are discussed below<br />

and shown in Table 3.3.5.<br />

In persons undergoing an elective splenectomy, vaccination should be completed,<br />

where possible, 2 weeks before the scheduled operation date. In an unplanned<br />

splenectomy, vaccination should commence approximately 1 week after the<br />

splenectomy has occurred. 143<br />

Children with splenic dysfunction should also be given antibiotic prophylaxis to<br />

prevent bacterial infection, until at least 5 years of age. 144,145 All asplenic persons<br />

and/or their parents/carers should also be educated about the importance<br />

of early investigation and treatment of febrile illnesses, including the use of<br />

emergency antibiotics. Asplenic persons are recommended to wear a medical<br />

alert. Vaccination cannot provide protection against all bacterial infections,<br />

or even all pneumococcal serotypes that cause IPD, hence it is particularly<br />

important that persons with asplenia are informed of the life-long increased risk<br />

of severe bacterial infection, even if they have been appropriately vaccinated.<br />

Pneumococcal vaccination<br />

Additional doses of pneumococcal vaccine are recommended for persons with<br />

asplenia, depending on their age and previous immunisation history, as shown<br />

in Table 3.3.5. Detailed information is provided in 4.13 Pneumococcal disease and<br />

in Table 2.1.11 Catch-up schedule for 13vPCV (Prevenar 13) and 23vPPV (Pneumovax<br />

23) in children with a medical condition(s) associated with an increased risk of IPD,<br />

presenting at age


4vMenCV is recommended from 12 months of age, instead of the routine NIPscheduled<br />

MenCCV vaccination; a 2nd dose of 4vMenCV should be given at<br />

least 8 weeks later. (See also 4.10 Meningococcal disease.) For adults and children<br />

who are >12 months of age at diagnosis, 2 doses of 4vMenCV are recommended,<br />

a minimum of 8 weeks apart. Subsequent booster doses of 4vMenCV are<br />

recommended at 5-yearly intervals thereafter.<br />

Hib vaccination<br />

A single dose of Hib vaccine is recommended for asplenic persons who were not<br />

vaccinated in infancy or who are incompletely vaccinated (see 4.3 Haemophilus<br />

influenzae type b and Table 2.1.8 Catch-up schedule for Hib vaccination for children<br />


Table 3.3.5: Recommendations for vaccination in persons with functional or<br />

anatomical asplenia<br />

Age Recommendations<br />

Pneumococcal vaccines<br />

6 weeks to 5 to


Age Recommendations<br />

Haemophilus influenzae type b (Hib) vaccine<br />

6 weeks–


Persons with autoimmune diseases and other chronic conditions<br />

Persons with autoimmune conditions, such as systemic lupus erythematosus<br />

(SLE), rheumatoid arthritis (RA) and multiple sclerosis (MS), are at higher<br />

risk of infections, including vaccine-preventable diseases, with the associated<br />

potential morbidity and mortality from infection. <strong>The</strong>y are also at risk of<br />

infection due to treatment with immunosuppressive agents such as bDMARDs<br />

and targeted biological therapies. 148 Some diseases can be reactivated during<br />

therapy, so screening for infections such as hepatitis B and tuberculosis should be<br />

undertaken prior to vaccination. 149,150<br />

Overall, theoretical concerns that vaccines exacerbate or cause autoimmune<br />

diseases such as rheumatoid arthritis, type 1 diabetes and multiple sclerosis<br />

have not been substantiated, with sporadic case reports not verified by larger<br />

epidemiological studies. 151-154 However, persons with a history of Guillain-Barré<br />

syndrome (GBS) have an increased likelihood in general of developing GBS<br />

again, and the chance of them coincidentally developing the syndrome following<br />

influenza vaccination may be higher than in persons with no history of GBS.<br />

A small increased risk of GBS was associated historically with one influenza<br />

vaccine in the United States in 1976, but, since then, close surveillance has shown<br />

that GBS has occurred at a very low rate of up to 1 in 1 million doses of influenza<br />

vaccine, if at all. 155<br />

In persons with autoimmune diseases and other chronic conditions,<br />

there is potential for reduced immunogenicity of vaccines, due to both<br />

immunosuppressive therapies and the underlying disease. 156-158 <strong>The</strong> duration<br />

of immunocompromise may be prolonged and caution must be taken when<br />

considering live vaccines. Inactivated vaccines are recommended, to optimise<br />

protection despite the potential for reduced immunogenicity in some people.<br />

Clinical and laboratory measures of disease activity, and the choice, duration<br />

and dose of immunosuppressive therapies, do not always predict who will<br />

respond poorly to vaccination. 157,159,160 In some instances, due to ongoing risk<br />

of disease, additional vaccine doses may be required, such as pneumococcal<br />

vaccine. Inactivated vaccines such as HPV and dTpa can be administered to<br />

immunocompromised persons. Annual influenza vaccine is also important in<br />

this population and should be administered annually (2 doses in the first year,<br />

1 annually thereafter).<br />

Hypopituitarism is not a contraindication to vaccination if the person is only<br />

receiving physiological corticosteroid replacement, as this is not considered<br />

immunosuppressive. If the person has been unwell and is on high-dose<br />

corticosteroids for more than 1 week, the use of live attenuated vaccines should<br />

be delayed for a minimum of 1 month.<br />

Persons with metabolic diseases should be vaccinated using the routine schedule,<br />

as vaccinations are generally considered safe in these persons. 161 Influenza and<br />

pneumococcal vaccines are recommended for those with metabolic disease. Any<br />

individual concerns should be discussed with the treating metabolic physician.<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 165<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS


3.3.4 Vaccination of recent recipients of normal human<br />

immunoglobulin and other blood products<br />

<strong>The</strong> immune response to live parenteral viral vaccines (with the exception<br />

of yellow fever and zoster vaccine) may be inhibited by normal human<br />

immunoglobulin (NHIG). <strong>The</strong> interval recommended is dependent on the type<br />

and half-life of the immunoglobulin administered (see Table 3.3.6 Recommended<br />

intervals between either immunoglobulins or blood products and MMR, MMRV or<br />

varicella vaccination).<br />

Rotavirus vaccine may be administered at any time before or after, or<br />

concurrently with, any blood product, including antibody-containing products,<br />

following the routinely recommended schedule for rotavirus vaccine among<br />

infants who are eligible for vaccination (see 4.17 Rotavirus). Minimal data are<br />

available on the impact of blood products on the immune response to the vaccine<br />

in these infants. Completing the full rotavirus vaccine series will optimise<br />

protection. 162<br />

Zoster vaccine can be given at any time before or after administration of<br />

immunoglobulin, or any antibody-containing blood product, because those<br />

for whom it is registered (persons ≥50 years of age) are assumed to have had<br />

a previous VZV infection and, therefore, already have serum antibody levels<br />

comparable to those found in blood products (see 4.24 Zoster).<br />

In persons with agammaglobulinaemia who are receiving monthly NHIG,<br />

the use of live vaccines is not recommended as the immune response may be<br />

inhibited. In addition, these people will have sufficient circulating antibody<br />

(e.g. measles, varicella) from the NHIG to protect them in the case of exposure.<br />

Inactivated vaccines are recommended as per the routine schedule; the response<br />

may be suboptimal, but these vaccines are safe to administer.<br />

Persons, who have received a blood transfusion, including mass blood<br />

transfusions, do not require any past vaccinations to be repeated. However,<br />

following the receipt of any blood product, including plasma or platelets, an<br />

interval of 3 to 11 months should elapse, dependent on the blood product<br />

transfused, before vaccination with an MMR, MMRV or varicella vaccine (see<br />

Table 3.3.6 Recommended intervals between either immunoglobulins or blood products<br />

and MMR, MMRV or varicella vaccination). An interval is suggested because there<br />

may be low levels of antibodies present in the blood product that may impair the<br />

immune response to the live vaccine.<br />

166 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Table 3.3.6: Recommended intervals between either immunoglobulins or blood<br />

products and measles-mumps-rubella (MMR), measles-mumpsrubella-varicella<br />

(MMRV) or varicella vaccination* 163<br />

Immunoglobulin/blood product Route Dose Interval<br />

(months)<br />

IU or mL Estimated<br />

mg IgG/kg<br />

Blood transfusion:<br />

Washed RBCs<br />

IV<br />

10 mL/kg<br />

Negligible<br />

RBCs, adenine-saline added IV 10 mL/kg 10 3<br />

Packed RBCs IV 10 mL/kg 20–60 5<br />

Whole blood IV 10 mL/kg 80–100 6<br />

Cytomegalovirus immunoglobulin IV 3 mL/kg 150 6<br />

HBIG as hepatitis B prophylaxis IM 100 IU<br />

400 IU<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 167<br />

0<br />

10 3<br />

NHIG (intravenous) for ITP treatment IV 400 8<br />

NHIG (intravenous) for ITP treatment IV 1000 10<br />

NHIG (intravenous) for ITP or<br />

Kawasaki disease treatment<br />

IV 1600–2000 11<br />

NHIG as hepatitis A prophylaxis IM 0.5 mL (50 kg)<br />

3<br />

NHIG as measles prophylaxis:<br />

(max. dose 15 mL)<br />

Standard<br />

IM 0.2 mL/kg<br />

5<br />

Immunocompromised IM 0.5 mL/kg<br />

6<br />

Plasma or platelet products IV 10 mL/kg 160 7<br />

HRIG as rabies prophylaxis IM 20 IU/kg 22 4<br />

Replacement (or therapy) of immune<br />

deficiencies (as NHIG [intravenous],<br />

various doses)<br />

IV 300–400 9<br />

Rh (D) IG (anti-D) IM 0<br />

TIG (IM use) for tetanus prophylaxis IM 250 IU (given<br />

within 24 hours<br />

of injury)<br />

500 IU (>24 hours<br />

after injury)<br />

ZIG as varicella prophylaxis IM 200 IU (0–10 kg)<br />

400 IU (11–30 kg)<br />

600 IU (>30 kg)<br />

* Zoster vaccine can be given at any time before or after administration of immunoglobulin or any<br />

antibody-containing blood product.<br />

10<br />

20<br />

3<br />

5<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS


3.3.5 Vaccination of persons with bleeding disorders<br />

Persons who are receiving anticoagulant therapy may develop haematomas in<br />

IM injection sites. <strong>The</strong> length of anticoagulant therapy should be clarified and<br />

immunisation delayed if therapy is going to be of short-term duration. Unless<br />

warfarin or low molecular weight heparin (LMWH) doses are known to be<br />

stable, persons receiving anticoagulants should have appropriate levels checked<br />

before vaccine administration, if possible. Intramuscular injections should be<br />

deferred if the INR is >2.0 (warfarin) or the anti-Xa (LMWH) level 4 hours post<br />

dose is >0.5 Units/mL.<br />

If a person has haemophilia and is receiving clotting factor replacement or similar<br />

therapy, IM vaccine administration should be conducted as soon as possible after<br />

the medication is received. 163 <strong>The</strong> site should not be rubbed post administration,<br />

but firm pressure applied for approximately 5–10 minutes. Vaccine recipients<br />

and/or carers should be informed about the possibility of haematoma formation.<br />

Ice and immobilisation may be used in the case of a small haematoma. <strong>The</strong><br />

subcutaneous route could be considered as an alternative in a person with<br />

haemophilia or on anticoagulant therapy; however, the intramuscular route is<br />

preferred if that is the usual recommended mode of vaccine administration –<br />

seek expert advice. If a vaccine is administered subcutaneously, there may be<br />

diminished immune response (e.g. requirement to check anti-HBs antibodies)<br />

and additional vaccine doses may be required. 164,165<br />

3.3.6 Vaccination before or after anaesthesia/surgery<br />

Recent or imminent surgery is not a contraindication to vaccinations, and<br />

recent vaccination is not a contraindication to surgery (see 2.1.4 Pre-vaccination<br />

screening). <strong>The</strong>re are no randomised controlled trials providing evidence of<br />

adverse outcomes with anaesthesia and surgery in recently vaccinated children.<br />

It is possible that the systemic effects from recent vaccination, such as fever and<br />

malaise, may cause confusion in the post-operative period. As the evidence<br />

is limited, it is possible to administer vaccines as per the routine schedule,<br />

or electively during a procedure for a person in a special risk group, if the<br />

appropriate vaccine delivery safety mechanisms are in place. 166<br />

If elective surgery and anaesthesia are to be postponed, some guidelines<br />

recommend postponing for 1 week after inactive vaccination and for 3 weeks<br />

after live attenuated viral vaccination in children. Routine vaccination may be<br />

deferred for 1 week after surgery. 167<br />

A person who receives any blood products during surgery will need to be<br />

informed of the need to delay some vaccinations (see Table 3.3.6 Recommended<br />

intervals between either immunoglobulins or blood products and MMR, MMRV or<br />

varicella vaccination).<br />

168 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


3.3.7 Vaccination of persons at occupational risk<br />

Certain occupations, particularly those associated with healthcare, are associated<br />

with an increased risk of some vaccine-preventable diseases. 168,169 Furthermore,<br />

some infected workers, particularly healthcare workers and those working in<br />

early childhood education and care, may transmit infections such as influenza,<br />

rubella, measles, mumps, varicella and pertussis to susceptible contacts, with<br />

the potential for serious health outcomes. Many infectious diseases, measles in<br />

particular, are highly infectious several days before symptoms become apparent.<br />

Healthcare workers employed within the public health system should check<br />

local state or territory healthcare worker immunisation requirements and the<br />

necessary documentation required (see Appendix 1 Contact details for <strong>Australian</strong>,<br />

state and territory government health authorities and communicable disease control).<br />

Where workers are at significant occupational risk of acquiring a vaccinepreventable<br />

disease, the employer should implement a comprehensive<br />

occupational vaccination program, which includes a vaccination policy, current<br />

staff vaccination records, provision of information about the relevant vaccinepreventable<br />

diseases, and the management of vaccine refusal (e.g. reducing<br />

the risk of a healthcare worker transmitting disease to vulnerable persons).<br />

Employers should take all reasonable steps to encourage non-immune workers to<br />

be vaccinated.<br />

Current recommended vaccinations for persons at risk of occupationally acquired<br />

vaccine-preventable diseases are listed in Table 3.3.7. In addition to the vaccines<br />

specific to a person’s occupation and work-related activities recommended here,<br />

all adults should be up to date with routinely recommended vaccines, such as<br />

dT-containing and MMR vaccines. (See also Table 2.1.12 in 2.1.5 Catch-up.)<br />

Standard precautions should be adopted where there is risk of occupational<br />

exposure to blood and body fluids. Preventive measures include the appropriate<br />

handling and disposal of sharps, the donning of gloves when handling body<br />

fluids, and the use of goggles/face shields when splashes are likely.<br />

If a non-immune person is exposed to a vaccine-preventable disease, postexposure<br />

prophylaxis should be administered where indicated (see relevant<br />

disease-specific chapters in Part 4, and Part 5 Passive immunisation).<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 169<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS


Table 3.3.7: Recommended vaccinations for persons at increased risk of certain<br />

occupationally acquired vaccine-preventable diseases* †<br />

Occupation Vaccine<br />

Healthcare workers (HCW)<br />

All HCW<br />

Includes all workers and students directly involved in<br />

patient care or the handling of human tissues<br />

HCW who work in remote Indigenous communities<br />

or with Indigenous children in NT, Qld, SA and<br />

WA, and other specified healthcare workers in some<br />

jurisdictions<br />

HCW who may be at high risk of exposure to drugresistant<br />

cases of tuberculosis (dependent on state or<br />

territory guidelines)<br />

Persons who work with children<br />

All persons working with children, including:<br />

• staff and students working in early childhood<br />

education and care<br />

• correctional staff working where infants/<br />

children cohabitate with mothers<br />

• school teachers (including student teachers)<br />

• outside school hours carers<br />

• child counselling services workers<br />

• youth services workers<br />

170 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Hepatitis B<br />

Influenza<br />

MMR (if non-immune) ‡<br />

Pertussis (dTpa)<br />

Varicella (if non-immune)<br />

Vaccines listed for ‘All<br />

HCW’, plus hepatitis A<br />

Vaccines listed for ‘All<br />

HCW’, plus consider BCG<br />

Influenza<br />

MMR (if non-immune) ‡<br />

Pertussis (dTpa)<br />

Varicella (if non-immune)<br />

Staff working in early childhood education and care Vaccines listed for ‘Persons<br />

who work with children’,<br />

plus hepatitis A<br />

Carers<br />

Carers of persons with developmental disabilities § Hepatitis A<br />

Hepatitis B<br />

Influenza<br />

Staff of nursing homes and long-term care facilities for<br />

persons of any age §<br />

Influenza<br />

MMR (if non-immune) ‡<br />

Varicella (if non-immune)


Occupation Vaccine<br />

Providers of home care to persons at risk of high Influenza<br />

influenza morbidity<br />

Emergency and essential service workers<br />

Police and emergency workers Hepatitis B<br />

Influenza<br />

Tetanus (dT or dTpa)<br />

Armed forces personnel Hepatitis B<br />

Influenza<br />

MMR (if non-immune) ‡<br />

Tetanus (dT or dTpa)<br />

Other vaccines relevant to<br />

deployment<br />

Staff of correctional facilities Hepatitis B<br />

Influenza<br />

MMR (if non-immune) ‡<br />

Tetanus (dT or dTpa)<br />

Staff of detention and immigration centres Hepatitis B<br />

Influenza<br />

MMR (if non-immune) ‡<br />

Tetanus (dT or dTpa)<br />

Laboratory personnel<br />

Laboratory personnel handling veterinary specimens Q fever<br />

or working with Q fever organism (Coxiella burnetii)<br />

Laboratory personnel handling either bat tissues or<br />

lyssaviruses (including rabies virus and <strong>Australian</strong> bat<br />

lyssavirus)<br />

Laboratory personnel routinely working with these<br />

organisms:<br />

Bacillus anthracis<br />

Vaccinia poxviruses<br />

Poliomyelitis virus<br />

Salmonella enterica subspecies enterica<br />

serovar Typhi (S. Typhi)<br />

Yellow fever virus<br />

Neisseria meningitidis<br />

Japanese encephalitis virus<br />

Rabies<br />

Anthrax <br />

Smallpox <br />

Poliomyelitis (IPV)<br />

Typhoid<br />

Yellow fever<br />

Quadrivalent<br />

meningococcal conjugate<br />

vaccine (4vMenCV)<br />

Japanese encephalitis<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 171<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS


Occupation Vaccine<br />

Persons who work with specific communities<br />

Workers who live with, or make frequent visits to,<br />

remote Indigenous communities in NT, Qld, SA and WA<br />

172 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Hepatitis A<br />

Workers assigned to the outer Torres Strait Islands for Japanese encephalitis<br />

a total of 30 days or more during the wet season<br />

Persons who work with animals<br />

Veterinarians, veterinary students, veterinary nurses # Influenza<br />

Q fever<br />

Rabies<br />

Agricultural college staff and students (aged >15<br />

years) exposed to high-risk animals #<br />

Q fever<br />

Abattoir workers and contract workers in abattoirs<br />

(excluding pig abattoirs)<br />

Livestock transporters<br />

Sheep shearers and cattle, sheep and dairy farmers<br />

Those culling or processing kangaroos or camels<br />

Tanning and hide workers<br />

Goat farmers<br />

Livestock saleyard workers<br />

Those handling animal products of conception<br />

Wildlife and zoo workers who have contact with atrisk<br />

animals, including kangaroos and bandicoots<br />

Persons who come into regular contact with bats<br />

(both ‘flying foxes’ and microbats), bat handlers, bat<br />

scientists, wildlife officers, zoo curators<br />

Q fever<br />

Q fever<br />

Rabies<br />

Poultry workers and others handling poultry,<br />

Influenza<br />

including those who may be involved in culling<br />

during an outbreak of avian influenza, and swine<br />

industry workers<br />

Other persons exposed to human tissue, blood, body fluids or sewage<br />

Embalmers Hepatitis B<br />

Workers who perform skin penetration procedures Hepatitis B<br />

(e.g. tattooists, body-piercers)<br />

Funeral workers and other workers who have regular<br />

contact with human tissue, blood or body fluids and/<br />

or used needles or syringes<br />

Plumbers or other workers in regular contact with<br />

untreated sewage<br />

Hepatitis B<br />

Hepatitis A<br />

Tetanus (dT or dTpa)<br />

* Work activities, rather than job title, should be considered on an individual basis to ensure an<br />

appropriate level of protection is afforded to each worker. In addition to providing protection


against certain vaccine-preventable diseases that persons in these occupations may be at<br />

increased risk of acquiring, vaccination may also reduce the risk of transmission of diseases to<br />

others with whom these persons are in contact.<br />

† In addition to the vaccines specific to a person’s occupation and work-related activities<br />

recommended here, all adults should be up to date with routinely recommended vaccines, such<br />

as dT-containing and MMR vaccines (see also Table 2.1.12 in 2.1.5 Catch-up).<br />

‡ All adults born during or since 1966 should have evidence of either receiving 2 doses of MMR<br />

vaccine or having immunity to measles, mumps and rubella. Adults born before 1966 are<br />

considered to be immune due to extensive measles, mumps and rubella circulating widely in<br />

the community during this period of time (see 4.9 Measles).<br />

§ Carers of infants


If a child is ≥12 months of age, the 1st doses of DTPa, hepatitis B, IPV, MMR,<br />

MenCCV, 13vPCV and Hib vaccines can be given at the same visit. For details,<br />

see 2.1.5 Catch-up.<br />

Migrant/refugee adults also need to be targeted for vaccination, especially<br />

against rubella, using MMR vaccine. This is particularly important for women<br />

of child-bearing age. Some refugees aged between 9 months and 30 years may<br />

have been offered MMR as part of a pre-departure screening, but may require a<br />

subsequent dose on arrival in Australia. 170 It is important to take into account any<br />

live attenuated viral vaccines that may have been administered as part of a predeparture<br />

screening, such as measles-containing vaccines or yellow fever vaccine<br />

(especially in those persons arriving from central and northern African nations).<br />

It is important to allow a minimum 4-week interval before administering any<br />

other live attenuated viral vaccines.<br />

All vaccines administered to children


3.3.11 Vaccination of persons who inject drugs<br />

Persons who inject drugs are at risk of acquiring hepatitis A and hepatitis B,<br />

and should be vaccinated against these infections (see 4.4 Hepatitis A and<br />

4.5 Hepatitis B). In addition, persons who inject drugs should be up to date<br />

with routinely recommended vaccines for adults, such as dT-containing and<br />

MMR vaccines. (See also Table 2.1.12 in 2.1.5 Catch-up.)<br />

3.3.12 Vaccination of sex industry workers<br />

Sex industry workers are at risk of acquiring hepatitis A and hepatitis B,<br />

and should be vaccinated against these infections (see 4.4 Hepatitis A and<br />

4.5 Hepatitis B). Human papillomavirus vaccine may also be indicated<br />

(see 4.6 Human papillomavirus). In addition, sex industry workers should be<br />

up to date with routinely recommended vaccines for adults, such as dTcontaining<br />

and MMR vaccines. (See also Table 2.1.12 in 2.1.5 Catch-up.)<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

PART 3 VACCINATION FOR SPECIAL RISK GROUPS 175<br />

3.3 GROUPS WITH SPECIAL<br />

VACCINATION REQUIREMENTS


PART 4 VACCINE-PREVENTABLE DISEASES<br />

4.1 CHOLERA<br />

4.1.1 Bacteriology<br />

Vibrio cholerae is a motile, curved Gram-negative bacillus. Differences in the<br />

O antigens have led to the description of more than 150 serogroups, only two<br />

of which have been found to cause cholera. Cholera is caused by enterotoxinproducing<br />

V. cholerae of serogroups O1 and O139 (sometimes referred to as<br />

the ‘Bengal’ strain). Serogroup O1 includes two biotypes (classical and El Tor),<br />

each of which includes organisms of Inaba, Ogawa and Hikojima serotypes.<br />

<strong>The</strong> ability of V. cholerae to persist in water is determined by the temperature,<br />

pH, salinity and availability of nutrients; it can survive under unfavourable<br />

conditions in a viable dormant state. 1 Transmission predominantly occurs when<br />

people ingest faecally contaminated food or water.<br />

4.1.2 Clinical features<br />

Cholera is an acute bacterial infection that is generally characterised by the<br />

sudden onset of painless, profuse, watery diarrhoea. In rare situations more than<br />

half the severe cases will die. Mild cases also occur, as does subclinical infection. 1<br />

<strong>The</strong> cholera toxin does not produce intestinal inflammation. <strong>The</strong> cholera toxin<br />

induces secretion of increased amounts of electrolytes into the intestinal lumen,<br />

resulting in mild to severe dehydration and, in some cases, metabolic acidosis.<br />

4.1.3 Epidemiology<br />

<strong>The</strong> disease is usually transmitted via food and water contaminated with human<br />

excreta. Seafood such as shellfish obtained from contaminated waters have<br />

also been responsible for outbreaks. 1 Cholera is a substantial health burden in<br />

developing countries and is considered to be endemic in Africa, Asia, South<br />

America and Central America. 2 Cholera epidemics are common in circumstances<br />

where food and water supplies can become contaminated, such as after natural<br />

disasters and civil unrest. 2 Cases of cholera in Australia (about 2 to 6 cases a<br />

year) almost always occur in individuals who have been infected in endemic<br />

areas overseas. 3 However, the overall risk of cholera to travellers with access to<br />

a safe water source and hygienic food preparation is considered to be low, even<br />

when visiting countries where cholera is endemic. <strong>The</strong> risk of infection has been<br />

estimated at 0.2 cases per 100 000 travellers from western countries, and the risk<br />

of severe disease is considerably lower, 4 although under-detection and underreporting<br />

of cholera among travellers is likely. 2,4,5<br />

In 1977, a locally acquired case led to the discovery of V. cholerae in some rivers<br />

of the Queensland coast. 6 Because of this, health workers should be aware that<br />

sporadic cases of cholera may, on rare occasions, follow contact with estuarine<br />

176 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


waters. All cases of cholera reported since the commencement of the National<br />

Notifiable Diseases Surveillance System in 1991 have been acquired outside<br />

Australia, except for 1 case of laboratory-acquired cholera in 1996 and 3 cases in<br />

2006. 3,7 <strong>The</strong> 3 cases in 2006, reported in Sydney, were linked and associated with<br />

consumption of raw imported whitebait. 7 <strong>The</strong>se patients had no history of recent<br />

travel to known cholera-endemic areas. 7<br />

4.1.4 Vaccines<br />

• Dukoral – CSL Limited and Crucell Sweden AB (inactivated wholecell<br />

V. cholerae O1, in combination with a recombinant cholera toxin<br />

B subunit [rCTB]). Each 3.0 mL liquid vaccine dose vial contains heat<br />

and formalin-inactivated Inaba, Ogawa, classic and El Tor strains of<br />

V. cholerae O1, 31.25 x 10 9 vibrios of each, combined with 1.0 mg rCTB.<br />

<strong>The</strong> buffer consists of a sachet of effervescent granules of anhydrous<br />

sodium carbonate, sodium bicarbonate, anhydrous citric acid, sodium<br />

citrate, saccharin sodium and raspberry flavour. This formulation<br />

does not contain aspartame.<br />

Trials of the oral cholera vaccine that contained inactivated whole-cell V. cholerae<br />

O1 combined with rCTB have been performed mainly in Bangladesh and Peru. 8-14<br />

<strong>The</strong> large randomised controlled trial in Bangladesh included over 120 000<br />

children (aged 2–15 years) and women (aged >15 years), with up to 5 years<br />

follow-up. About 13 000 children and 8 000 women received 3 doses of the study<br />

vaccine. When cholera cases in all age groups were aggregated, the protective<br />

efficacy of this vaccine (in a 3-dose regimen with inactivated Escherichia coli as<br />

control) was 85%, 6 months after the 3rd dose. <strong>The</strong> protective efficacy decreased<br />

to 62% after 1 year, and to 57% after 2 years. 8,10 On long-term follow-up (up to<br />

5 years) no significant protective efficacy was observed beyond 2 years. 8,14 <strong>The</strong><br />

efficacy of the vaccine was lower and waned more rapidly in children aged<br />

2–5 years. 14 In this age group, while the efficacy was 100% during the first<br />

4–6 months after vaccination, it became non-significant in the latter half of the<br />

1st year of follow-up (during a cholera epidemic), resulting in an overall efficacy<br />

of 38% after 1 year; efficacy after 2 years was comparable. In contrast, for those<br />

aged >5 years, the efficacy estimates were 76%, 78% and 63%, respectively, at<br />

these three time points. 8,9,14 <strong>The</strong> protective efficacy of the vaccine, over a 3-year<br />

follow-up period, was not significantly different among those who received a<br />

total of 2 doses versus those who received 3 doses (including all ages). 8,9<br />

A randomised controlled trial in Peru among military recruits aged 16–45 years<br />

found a vaccine efficacy of 86% against symptomatic cholera after 2 vaccine<br />

doses. 13 Another Peruvian household study showed an overall efficacy of 61%<br />

PART 4 VACCINE-PREVENTABLE DISEASES 177<br />

4.1 CHOLERA


among 2–65-year olds, 12 after a booster dose given 10 months after a 2-dose<br />

primary series. 12 A field effectiveness case-control study in Mozambique, during<br />

a mass oral cholera vaccination program in an endemic population aged<br />

≥2 years, found that 1 or more doses of the inactivated oral cholera vaccine was<br />

78% protective (1–6 months after the 1st dose). <strong>The</strong> per-protocol effectiveness of<br />

2 doses was 84% (0.5–4.5 months after the 2nd dose). 15<br />

<strong>The</strong>re is structural similarity and immunologic cross-reactivity between the<br />

cholera toxin and the heat-labile toxin of E. coli, which is often associated with<br />

‘travellers’ diarrhoea’. <strong>The</strong>refore, it had been suggested that the rCTB-containing<br />

vaccine may also provide protection against heat-labile toxin producing<br />

enterotoxigenic E. coli (LT-ETEC). A study in short-term Finnish tourists16 showed that the inactivated oral cholera vaccine also provided a 60% reduction<br />

in diarrhoea caused by LT-ETEC. A study in Bangladesh, an endemic area,<br />

showed 67% protection against LT-ETEC for 3 months only. 17 It can be expected<br />

that the inactivated vaccine will reduce the proportion of travellers’ diarrhoea<br />

that is caused by LT-ETEC. Approximately 30 to 40% of travellers to developing<br />

countries contract travellers’ diarrhoea, with an average of 20% of cases caused<br />

by LT-ETEC; hence, the 60% efficacy of the oral inactivated vaccine against<br />

LT-ETEC could be expected to prevent up to 15% of travellers’ diarrhoea. 18-20<br />

However, in Australia this vaccine is only registered for the prevention of<br />

cholera.<br />

To date, there is no vaccine marketed in Australia to protect against infection<br />

with V. cholerae O139. An oral killed whole-cell bivalent cholera vaccine (against<br />

both serogroups O1 and O139) has been evaluated in Vietnam. 21,22 More recently,<br />

in India, an interim analysis of a cluster-randomised controlled trial reported a<br />

protective efficacy of 67% against V. cholerae O1 after 2 years. Specific efficacy<br />

against V. cholerae 0139 could not be assessed in this study, as cholera episodes<br />

caused by this serogroup were not detected. 23<br />

4.1.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 24 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

Because the person to be vaccinated will be responsible for looking after the<br />

vaccine following purchase, details of how it should be transported (from<br />

pharmacy to home) and stored in the refrigerator (at home) must be carefully<br />

explained.<br />

4.1.6 Dosage and administration<br />

Dukoral is an oral vaccine.<br />

Food and drink should be avoided for 1 hour before and 1 hour after<br />

administration of the inactivated cholera vaccine, as the vaccine is acid labile.<br />

178 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Children aged 2–6 years<br />

Three doses are required, given a minimum of 1 week and up to 6 weeks apart.<br />

If an interval of more than 6 weeks occurs between any of the doses, re-start the<br />

vaccination course.<br />

Dukoral is administered orally. After dissolving the buffer granules in 150 mL<br />

of water, half the solution is then poured away and the entire contents of the<br />

vaccine vial are mixed with the remaining 75 mL for administration.<br />

Adults and children aged >6 years<br />

Two doses are required, given a minimum of 1 week and up to 6 weeks apart. If<br />

the 2nd dose is not administered within 6 weeks, re-start the vaccination course.<br />

Dukoral is administered orally. After dissolving the buffer granules in 150 mL<br />

of water, the contents of the vaccine vial are then added to the solution for<br />

administration.<br />

Co-administration with other vaccines<br />

<strong>The</strong> inactivated oral cholera vaccine can be given with, or at any time before or<br />

after, other travel vaccines, such as yellow fever or parenteral Vi polysaccharide<br />

typhoid vaccines.<br />

However, there should be an interval of at least 8 hours between the<br />

administration of the inactivated oral cholera vaccine and oral live attenuated<br />

typhoid vaccine (see 4.1.10 Precautions below).<br />

4.1.7 Recommendations<br />

Vaccination against cholera is not an official requirement for entry into any<br />

foreign country.<br />

Routine cholera vaccination is not recommended as the risk to travellers is<br />

very low, despite the endemicity of cholera in some countries often visited by<br />

<strong>Australian</strong>s. Careful and sensible selection of food and water is of far greater<br />

importance to the traveller than cholera vaccination.<br />

Cholera vaccination should be considered for travellers at increased risk of<br />

acquiring diarrhoeal disease, such as those with achlorhydria, and for travellers<br />

at increased risk of severe or complicated diarrhoeal disease, such as those<br />

with poorly controlled or otherwise complicated diabetes, inflammatory bowel<br />

disease, HIV/AIDS or other conditions resulting in immunocompromise, or<br />

significant cardiovascular disease.<br />

Cholera vaccination should also be considered for travellers with considerable<br />

risk of exposure to, or acquiring, cholera, such as humanitarian disaster workers<br />

deployed to regions with endemic or epidemic cholera.<br />

Dukoral is not registered for use in children aged


Booster doses<br />

Booster doses are recommended for those who are at ongoing risk of exposure to<br />

cholera.<br />

Children aged 2–6 years who are at ongoing risk should receive a single booster<br />

dose 6 months after completion of the primary course. If the interval between<br />

primary immunisation and the booster dose is more than 6 months, primary<br />

immunisation must be repeated.<br />

Adults and children aged >6 years who are at ongoing risk should receive a<br />

single booster dose up to 2 years after completion of the primary course. If the<br />

interval between primary immunisation and the booster dose is more than<br />

2 years, primary immunisation must be repeated.<br />

4.1.8 Pregnancy and breastfeeding<br />

Cholera vaccine is not routinely recommended for pregnant or breastfeeding<br />

women.<br />

<strong>The</strong>re is limited information on the use of inactivated oral cholera vaccines<br />

during pregnancy and breastfeeding. 25<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.1.9 Contraindications<br />

<strong>The</strong> only absolute contraindications to cholera vaccine are:<br />

• anaphylaxis following a previous dose of the vaccine<br />

• anaphylaxis following any vaccine component.<br />

4.1.10 Precautions<br />

Postpone administration of cholera vaccine during either an acute febrile illness<br />

or acute gastrointestinal illness with persistent diarrhoea or vomiting, until<br />

recovered.<br />

Although the vaccine is not contraindicated in people who are<br />

immunocompromised, including those with HIV infection, data on effectiveness<br />

in this population are limited.<br />

<strong>The</strong>re should be an interval of at least 8 hours between the administration of the<br />

inactivated oral cholera vaccine and oral live attenuated typhoid vaccine, as the<br />

buffer in the cholera vaccine may affect the transit of the capsules of oral typhoid<br />

vaccine through the gastrointestinal tract.<br />

180 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.1.11 Adverse events<br />

<strong>The</strong> inactivated oral cholera vaccine has a good safety profile, with similar<br />

rates of adverse events reported among vaccine and placebo clinical trial<br />

participants. 12,16,25 Mild abdominal pain, discomfort and diarrhoea were reported<br />

in post-marketing surveillance at a frequency of 0.1–1%. 26<br />

4.1.12 Public health management of cholera<br />

Cholera is a notifiable and quarantinable disease in all states and territories<br />

in Australia.<br />

Further instructions about the public health management of cholera,<br />

including management of cases of cholera and their contacts, should be<br />

obtained from state/territory public health authorities (see Appendix 1<br />

Contact details for <strong>Australian</strong>, state and territory government health authorities and<br />

communicable disease control).<br />

4.1.13 Variations from product information<br />

<strong>The</strong> production information for Dukoral states that a booster dose is<br />

recommended for adults 2 years after the completion of the primary vaccine<br />

course if there is an ongoing risk of cholera. <strong>The</strong> ATAGI recommends that a<br />

booster dose is also recommended 2 years after the completion of the vaccine<br />

course for children >6 years of age if there is an ongoing risk of cholera.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

PART 4 VACCINE-PREVENTABLE DISEASES 181<br />

4.1 CHOLERA


4.2 DIPHTHERIA<br />

4.2.1 Bacteriology<br />

Diphtheria is an acute illness caused by toxigenic strains of Corynebacterium<br />

diphtheriae, a Gram-positive, non-sporing, non-capsulate bacillus. <strong>The</strong> exotoxin<br />

produced by C. diphtheriae acts locally on the mucous membranes of the<br />

respiratory tract or, less commonly, on damaged skin, to produce an adherent<br />

pseudomembrane. Systemically, the toxin acts on cells of the myocardium,<br />

nervous system and adrenals.<br />

4.2.2 Clinical features<br />

<strong>The</strong> incubation period is 2 to 5 days. <strong>The</strong> disease is communicable for up to<br />

4 weeks, but carriers may shed organisms for longer. Spread is by aerosol<br />

transmission or by direct contact with skin lesions or articles soiled by infected<br />

persons. <strong>The</strong> disease can involve almost any mucous membrane. Pharyngeal<br />

diphtheria, by far the commonest form of disease in the unimmunised, is<br />

characterised by an inflammatory exudate that forms a greyish or green<br />

membrane in the upper respiratory tract, which can cause acute severe<br />

respiratory obstruction. Life-threatening complications from diphtheria toxin<br />

include myocarditis and neuritis (usually affecting motor nerves). <strong>The</strong> casefatality<br />

rate in the last three decades has been reported as up to 16%. 1 Diphtheria<br />

antitoxin, which neutralises unbound toxin, was first used in the 1890s. Together<br />

with antibiotics, antitoxin is the mainstay of treatment for diphtheria, but this<br />

may not always be successful. <strong>The</strong> first death from diphtheria in Australia for<br />

over 20 years occurred in 2011 in an unvaccinated person. 2 Effective protection<br />

against diphtheria is only achieved by active immunisation with diphtheria<br />

toxoid-containing vaccines. 1,3<br />

4.2.3 Epidemiology<br />

In the early 1900s, diphtheria caused more deaths in Australia than any other<br />

infectious disease, but increasing use of diphtheria vaccines since World War II<br />

has led to its virtual disappearance. 4 <strong>The</strong> current epidemiology of diphtheria<br />

in Australia is similar to that in other developed countries. Almost all recent<br />

cases in the United Kingdom and the United States have been associated with<br />

imported infections. 5 In Australia, there have been two imported infections<br />

identified, one case in 2001 and one imported infection in 2011, resulting in<br />

two additional cases, including one death. 2,6 <strong>The</strong> 2011 fatal case of pharyngeal<br />

diphtheria occurred in an unvaccinated person infected by a friend who acquired<br />

diphtheria in a less developed country. 2<br />

182 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.2.4 Vaccines<br />

Diphtheria toxoid is available in Australia only in combination with tetanus and<br />

other antigens.<br />

<strong>The</strong> acronym DTPa, using capital letters, signifies child formulations of<br />

diphtheria, tetanus and acellular pertussis-containing vaccines. <strong>The</strong> acronym<br />

dTpa is used for formulations that contain substantially lesser amounts<br />

of diphtheria toxoid and pertussis antigens than child (DTPa-containing)<br />

formulations; dTpa vaccines are usually used in adolescents and adults.<br />

Diphtheria vaccination stimulates the production of antitoxin, which protects<br />

against the toxin produced by the organism. <strong>The</strong> immunogen is prepared by<br />

treating a cell-free preparation of toxin with formaldehyde, thereby converting it<br />

into the innocuous diphtheria toxoid. Diphtheria toxoid is usually adsorbed onto<br />

an adjuvant, either aluminium phosphate or aluminium hydroxide, to increase<br />

its immunogenicity. Antigens from Bordetella pertussis, in combination vaccines,<br />

also act as an effective adjuvant.<br />

<strong>The</strong> circulating levels of antitoxin required for protection from diphtheria are<br />

well described. Antitoxin levels of 0.1 IU/mL are associated<br />

with more certain and prolonged protection. 7 Complete immunisation induces<br />

protective levels of antitoxin lasting throughout childhood, but, by middle age,<br />

at least 50% of persons not vaccinated since childhood have levels


• Infanrix IPV – GlaxoSmithKline (DTPa-IPV; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus). Each 0.5 mL pre-filled<br />

syringe contains ≥30 IU diphtheria toxoid, ≥40 IU tetanus toxoid,<br />

25 µg PT, 25 µg FHA, 8 µg PRN, 40 D-antigen units inactivated<br />

poliovirus type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1)<br />

and 32 D-antigen units type 3 (Saukett), adsorbed onto aluminium<br />

hydroxide; traces of formaldehyde, polysorbate 80, polymyxin and<br />

neomycin.<br />

• Pediacel – Sanofi Pasteur Pty Ltd (DTPa-IPV-Hib; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus-Haemophilus influenzae type<br />

b). Each 0.5 mL monodose vial contains ≥30 IU diphtheria toxoid,<br />

≥40 IU tetanus toxoid, 20 µg PT, 20 µg FHA, 3 µg PRN, 5 µg pertussis<br />

fimbriae (FIM) 2+3, 40 D-antigen units inactivated poliovirus type<br />

1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32 D-antigen<br />

units type 3 (Saukett), 10 µg Hib capsular polysaccharide conjugated<br />

to 20 µg tetanus protein; 1.5 mg aluminium phosphate; ≤50 ng<br />

bovine serum albumin; phenoxyethanol as preservative; traces of<br />

formaldehyde, glutaraldehyde, polysorbate 80, polymyxin, neomycin<br />

and streptomycin.<br />

• Quadracel – Sanofi Pasteur Pty Ltd (DTPa-IPV; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus). Each 0.5 mL monodose vial<br />

contains ≥30 IU diphtheria toxoid, ≥40 IU tetanus toxoid, 20 µg PT,<br />

20 µg FHA, 3 µg PRN, 5 µg FIM 2+3, 40 D-antigen units inactivated<br />

poliovirus type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and<br />

32 D-antigen units type 3 (Saukett); 1.5 mg aluminium phosphate;<br />

≤50 ng bovine serum albumin; phenoxyethanol as preservative; traces<br />

of formaldehyde, glutaraldehyde, polysorbate 80, polymyxin and<br />

neomycin.<br />

• Tripacel – Sanofi Pasteur Pty Ltd (DTPa; diphtheria-tetanus-acellular<br />

pertussis). Each 0.5 mL monodose vial contains ≥30 IU diphtheria<br />

toxoid, ≥40 IU tetanus toxoid, 10 µg PT, 5 µg FHA, 3 µg PRN, 5 µg<br />

FIM 2+3; 1.5 mg aluminium phosphate; 3.4 mg phenoxyethanol.<br />

Reduced antigen formulations for adults, adolescents and<br />

children aged ≥10 years<br />

• ADT Booster – CSL Limited/Statens Serum Institut (dT; diphtheriatetanus).<br />

Each 0.5 mL monodose vial or pre-filled syringe contains<br />

≥2 IU diphtheria toxoid and ≥20 IU tetanus toxoid, adsorbed onto<br />

0.5 mg aluminium as aluminium hydroxide.<br />

184 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


• Adacel – Sanofi Pasteur Pty Ltd (dTpa; diphtheria-tetanus-acellular<br />

pertussis). Each 0.5 mL monodose vial contains ≥2 IU diphtheria<br />

toxoid, ≥20 IU tetanus toxoid, 2.5 µg PT, 5 µg FHA, 3 µg PRN,<br />

5 µg FIM 2+3; 0.33 mg aluminium as aluminium phosphate;<br />

phenoxyethanol as preservative; traces of formaldehyde and<br />

glutaraldehyde.<br />

• Adacel Polio – Sanofi Pasteur Pty Ltd (dTpa-IPV; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus). Each 0.5 mL monodose vial<br />

or pre-filled syringe contains ≥2 IU diphtheria toxoid, ≥20 IU tetanus<br />

toxoid, 2.5 µg PT, 5 µg FHA, 3 µg PRN, 5 µg FIM 2+3, 40 D-antigen<br />

units inactivated poliovirus type 1 (Mahoney), 8 D-antigen units type<br />

2 (MEF-1) and 32 D-antigen units type 3 (Saukett); 0.33 mg aluminium<br />

as aluminium phosphate; phenoxyethanol as preservative; traces of<br />

formaldehyde, glutaraldehyde, polysorbate 80, polymyxin, neomycin<br />

and streptomycin.<br />

• Boostrix – GlaxoSmithKline (dTpa; diphtheria-tetanus-acellular<br />

pertussis). Each 0.5 mL monodose vial or pre-filled syringe contains<br />

≥2 IU diphtheria toxoid, ≥20 IU tetanus toxoid, 8 µg PT, 8 µg FHA,<br />

2.5 µg PRN, adsorbed onto 0.5 mg aluminium as aluminium<br />

hydroxide/phosphate; traces of formaldehyde, polysorbate 80 and<br />

glycine.<br />

• Boostrix-IPV – GlaxoSmithKline (dTpa-IPV; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus). Each 0.5 mL pre-filled<br />

syringe contains ≥2 IU diphtheria toxoid, ≥20 IU tetanus toxoid, 8 µg<br />

PT, 8 µg FHA, 2.5 µg PRN, 40 D-antigen units inactivated poliovirus<br />

type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32 D-antigen<br />

units type 3 (Saukett), adsorbed onto 0.5 mg aluminium as aluminium<br />

hydroxide/phosphate; traces of formaldehyde, polysorbate 80,<br />

polymyxin and neomycin.<br />

4.2.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 13 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

Infanrix hexa must be reconstituted by adding the entire contents of the syringe<br />

to the vial and shaking until the pellet is completely dissolved. Reconstituted<br />

vaccine should be used as soon as practicable. If storage is necessary, hold at<br />

room temperature for not more than 8 hours.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 185<br />

4.2 DIPHTHERIA


4.2.6 Dosage and administration<br />

<strong>The</strong> dose of all diphtheria-containing vaccines is 0.5 mL, to be given by<br />

IM injection.<br />

Do not mix DTPa- or dTpa-containing vaccines or dT vaccine with any other<br />

vaccine in the same syringe, unless specifically registered for use in this way.<br />

4.2.7 Recommendations<br />

Infants and children<br />

Diphtheria toxoid is given in combination with tetanus toxoid and acellular<br />

pertussis as DTPa vaccine. <strong>The</strong> recommended 3-dose primary schedule is at 2,<br />

4 and 6 months of age. <strong>The</strong> 1st dose can be given as early as 6 weeks of age, due<br />

to the high morbidity and occasional mortality associated with pertussis in very<br />

young infants. If the 1st dose is given at 6 weeks of age, the next scheduled doses<br />

should still be given at 4 months and 6 months of age (see 4.12 Pertussis).<br />

A booster dose of diphtheria-containing vaccine, usually provided as DTPa-<br />

IPV, is recommended at 4 years of age, but can be given as early as 3.5 years.<br />

For this booster dose, all brands of DTPa-containing vaccines are considered<br />

interchangeable.<br />

Where required, DTPa-containing vaccines can be given for catch-up for either<br />

the primary doses or booster dose in children aged


antibodies at an age when waning of diphtheria and tetanus immunity is<br />

commencing in the <strong>Australian</strong> population. 11 Diphtheria can be a significant risk<br />

for travellers to some countries (particularly Southeast Asia, New Guinea, the<br />

states of the former Soviet Union, Baltic countries or eastern European countries).<br />

Travellers to countries where health services are difficult to access should be<br />

adequately protected against diphtheria before departure. <strong>The</strong>y should receive a<br />

booster dose of dT (or dTpa if not given previously) if more than 10 years have<br />

elapsed since the last dose of dT-containing vaccine.<br />

For persons undertaking high-risk travel, consider giving a booster dose of either<br />

dTpa or dT (as appropriate) if more than 5 years have elapsed since the last dose<br />

of a dT-containing vaccine.<br />

Primary vaccination<br />

Persons who have not received any diphtheria vaccines are also likely to<br />

have missed tetanus vaccination. <strong>The</strong>refore, 3 doses of dT should be given at<br />

minimum intervals of 4 weeks, followed by booster doses at 10 and 20 years after<br />

the primary course. One of these 3 doses (preferably the 1st) should be given as<br />

dTpa, to also provide additional protection against pertussis. In the event that<br />

dT vaccine is not available, dTpa can be used for all primary doses. However,<br />

this is not recommended routinely because there are no data on the safety,<br />

immunogenicity or efficacy of dTpa in multiple doses for primary vaccination.<br />

For additional information on adults with no history of a primary course of dT<br />

vaccine requiring catch-up, see 2.1.5 Catch-up.<br />

4.2.8 Pregnancy and breastfeeding<br />

Although dT or dTpa vaccines are not routinely recommended for pregnant<br />

women, they can be given under certain circumstances, such as for management<br />

of a tetanus-prone wound (see 4.19 Tetanus) or to prevent pertussis in pregnant<br />

women and their newborns (see 4.12 Pertussis). 14<br />

dT or dTpa vaccines can be given to breastfeeding women.<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.2.9 Contraindications<br />

<strong>The</strong> only absolute contraindications to diphtheria-containing vaccines are:<br />

• anaphylaxis following a previous dose of any diphtheria-containing vaccine<br />

• anaphylaxis following any vaccine component.<br />

4.2.10 Adverse events<br />

Mild discomfort or pain at the injection site persisting for up to a few days is<br />

common. Uncommon general adverse events following dT vaccine include<br />

headache, lethargy, malaise, myalgia and fever. Anaphylaxis, urticaria and<br />

peripheral neuropathy occur very rarely. Brachial neuritis (inflammation of a<br />

PART 4 VACCINE-PREVENTABLE DISEASES 187<br />

4.2 DIPHTHERIA


nerve in the arm, causing weakness or numbness) has been described following<br />

the administration of tetanus toxoid-containing vaccines, with an estimated<br />

excess risk of approximately 0.5–1 in 100 000 doses in adults. 15,16 For specific<br />

adverse events following combination vaccines containing both diphtheria and<br />

pertussis antigens, see 4.12 Pertussis.<br />

4.2.11 Public health management of diphtheria<br />

Diphtheria is a notifiable disease in all states and territories in Australia.<br />

Further instructions about the public health management of diphtheria,<br />

including management of cases of diphtheria and their contacts, should<br />

be obtained from state/territory public health authorities (see Appendix 1<br />

Contact details for <strong>Australian</strong>, state and territory government health authorities and<br />

communicable disease control).<br />

Confirmed or suspected diphtheria is of considerable public health importance<br />

and should be notified immediately to state/territory public health authorities.<br />

In general, contacts of a proven or presumptive diphtheria case will require<br />

vaccination (either primary or booster, depending on vaccination status), and<br />

appropriate prophylactic antibiotics17 (see 4.12 Pertussis).<br />

Advice should be sought with respect to diphtheria antitoxin access and<br />

dosage, and special arrangements made if hypersensitivity is suspected; this<br />

can be coordinated through the relevant state/territory health authority (see<br />

Appendix 1 Contact details for <strong>Australian</strong>, state and territory government health<br />

authorities and communicable disease control and Part 5 Passive immunisation).<br />

4.2.12 Variations from product information<br />

<strong>The</strong> product information for Infanrix hexa states that this vaccine is indicated<br />

for primary immunisation of infants from the age of 6 weeks. <strong>The</strong> ATAGI<br />

recommends that this vaccine may also be used for catch-up of the primary<br />

schedule in children


children aged


<strong>The</strong> product information for Boostrix, Boostrix-IPV, Infanrix hexa and<br />

Infanrix IPV states that these vaccines are contraindicated in children with<br />

encephalopathy of unknown aetiology or with neurologic complications<br />

occurring within 7 days following a vaccine dose. <strong>The</strong> ATAGI recommends<br />

instead that the only contraindication is a history of anaphylaxis to a previous<br />

dose or to any of the vaccine components.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

190 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.3 HAEMOPHILUS INFLUENZAE TYPE B<br />

4.3.1 Bacteriology<br />

Haemophilus influenzae is a Gram-negative coccobacillus that is a normal part<br />

of upper respiratory tract flora. It can be isolated in two forms: capsular and<br />

non-capsular. Strains isolated from respiratory tract specimens, such as sputum<br />

and middle ear or sinus fluid, usually do not have a capsule, and are known<br />

as non-typeable Haemophilus influenzae (NTHi). Six capsular types (a to f) have<br />

been described and, before the introduction of vaccination against Haemophilus<br />

influenzae type b (Hib), almost all H. influenzae isolates from sterile sites (blood,<br />

cerebrospinal fluid, joint or pleural fluid) were of the b capsular type. 1<br />

Before Hib immunisation, invasive disease caused by Hib rarely occurred<br />

after the age of 5 years. This was because the prevalence of antibody to Hib<br />

progressively increased from the age of 2 years, thought to be related to exposure<br />

to Hib (or cross-reacting organisms) colonising the nasopharynx or other sites.<br />

Children


population. 5-7 <strong>The</strong> reduction in the incidence of Hib disease following routine<br />

vaccination has been particularly marked in Indigenous children, although<br />

absolute rates remain substantially higher than those in the non-Indigenous<br />

population. 8-10 Similar impressive reductions in Hib disease have been seen in<br />

other countries with routine childhood vaccination. 11,12 Since Hib disease has<br />

become relatively rare, cases of epiglottitis can no longer be assumed to be due<br />

to H. influenzae type b and, moreover, even when H. influenzae is isolated from<br />

a normally sterile site, it may not be type b. Thus, laboratory confirmation of<br />

H. influenzae infection and serotype should always be sought before vaccination<br />

failure is assumed. 13,14<br />

4.3.4 Vaccines<br />

Four types of conjugate Hib vaccines have been developed, each containing the<br />

Hib capsular polysaccharide polyribosylribitol phosphate (PRP) conjugated to a<br />

different carrier protein. Of these, PRP-OMP (conjugated to the outer membrane<br />

protein of Neisseria meningitidis), PRP-T (conjugated to tetanus toxoid) and<br />

HbOC (conjugated to a mutant diphtheria toxoid) elicit antibody responses<br />

associated with protection of children against Hib. <strong>The</strong> fourth vaccine type,<br />

PRP-D (conjugated to diphtheria toxoid), was less immunogenic and found to be<br />

poorly protective in high-risk populations, such as Indigenous children. 15 PRP-T<br />

has subsequently been included in a number of combination vaccines, including<br />

DTPa-hepB-IPV-Hib and Hib-MenCCV.<br />

In Australia, the differing epidemiology of invasive Hib disease by ethnicity<br />

and region has determined the recommendations for Hib vaccine choice (see 3.1<br />

Vaccination for Aboriginal and Torres Strait Islander people). <strong>The</strong>re have been four<br />

distinct eras of implementation of the Hib vaccination program for <strong>Australian</strong><br />

children, which are described in detail elsewhere. 10<br />

Some Hib combination vaccines containing acellular pertussis are known to<br />

produce lower Hib antibody responses than similar formulations containing<br />

whole-cell pertussis. 16 When administered according to the United Kingdom’s<br />

schedule as 3 primary doses at 2, 3 and 4 months of age without a booster, their<br />

use has been associated with an increased risk of vaccine failure. 17 In other<br />

European countries that routinely give a 4th dose around the time of the 1st<br />

birthday, as Australia does, no loss of effectiveness has been observed. 18,19<br />

Monovalent Hib vaccines<br />

• Act-HIB – Sanofi Pasteur Pty Ltd (PRP-T). Lyophilised powder<br />

in a monodose vial with a pre-filled diluent syringe. Each 0.5 mL<br />

reconstituted dose contains 10 µg Hib capsular polysaccharide<br />

(polyribosyl-ribitol-phosphate [PRP]) conjugated to 18–30 µg<br />

tetanus protein.<br />

192 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


• Hiberix – GlaxoSmithKline (PRP-T). Lyophilised pellet in a monodose<br />

vial with separate diluent. Each 0.5 mL reconstituted dose contains<br />

10 µg Hib capsular polysaccharide (PRP) conjugated to 30 µg tetanus<br />

toxoid; lactose.<br />

• Liquid PedvaxHIB – CSL Limited/Merck & Co Inc (PRP-OMP). Each<br />

0.5 mL monodose vial contains 7.5 µg Hib capsular polysaccharide<br />

(PRP) conjugated to 125 µg Neisseria meningitidis outer membrane<br />

protein (OMP) complex; 225 µg aluminium as aluminium hydroxide;<br />

35 µg borax.<br />

Combination vaccines that contain Hib<br />

• Infanrix hexa – GlaxoSmithKline (DTPa-hepB-IPV-Hib; diphtheriatetanus-acellular<br />

pertussis-hepatitis B-inactivated poliovirus-<br />

Haemophilus influenzae type b [PRP-T]). <strong>The</strong> vaccine consists of both<br />

a 0.5 mL pre-filled syringe containing ≥30 IU diphtheria toxoid,<br />

≥40 IU tetanus toxoid, 25 µg pertussis toxoid (PT), 25 µg filamentous<br />

haemagglutinin (FHA), 8 µg pertactin (PRN), 10 µg recombinant<br />

HBsAg, 40 D-antigen units inactivated poliovirus type 1 (Mahoney),<br />

8 D-antigen units type 2 (MEF-1) and 32 D-antigen units type 3<br />

(Saukett), adsorbed onto aluminium hydroxide/phosphate; traces<br />

of formaldehyde, polysorbate 80, polysorbate 20, polymyxin and<br />

neomycin; and a vial containing a lyophilised pellet of 10 µg purified<br />

Hib capsular polysaccharide (PRP) conjugated to 20–40 µg tetanus<br />

toxoid. May contain yeast proteins.<br />

• Menitorix – GlaxoSmithKline (Hib-MenCCV; Haemophilus influenzae<br />

type b [PRP-T]-meningococcal serogroup C–tetanus toxoid conjugate).<br />

Lyophilised powder in a monodose vial with a pre-filled diluent<br />

syringe. Each 0.5 mL reconstituted dose contains 5 µg Hib capsular<br />

polysaccharide (PRP) conjugated to 12.5 µg tetanus toxoid, and 5 µg<br />

Neisseria meningitidis serogroup C polysaccharide conjugated to 5 µg<br />

tetanus toxoid; traces of trometamol and sucrose.<br />

• Pediacel – Sanofi Pasteur Pty Ltd (DTPa-IPV-Hib; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus-Haemophilus influenzae type<br />

b [PRP-T]). Each 0.5 mL monodose vial contains ≥30 IU diphtheria<br />

toxoid, ≥40 IU tetanus toxoid, 20 µg PT, 20 µg FHA, 3 µg PRN, 5 µg<br />

pertussis fimbriae (FIM) 2+3, 40 D-antigen units inactivated poliovirus<br />

type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and<br />

32 D-antigen units type 3 (Saukett), 10 µg Hib capsular polysaccharide<br />

(PRP) conjugated to 20 µg tetanus protein; 1.5 mg aluminium<br />

phosphate; ≤50 ng bovine serum albumin; phenoxyethanol as<br />

preservative; traces of formaldehyde, glutaraldehyde, polysorbate 80,<br />

polymyxin, neomycin and streptomycin.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 193<br />

4.3 HAEMOPHILUS<br />

INFLUENZAE TYPE B


4.3.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 20 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

Act-HIB must be reconstituted by adding the entire contents of the diluent syringe<br />

to the vial and shaking until the powder is completely dissolved. Reconstituted<br />

vaccine must be used immediately.<br />

Hiberix must be reconstituted by adding the entire contents of the diluent container<br />

to the vial and shaking until the pellet is completely dissolved. Reconstituted<br />

vaccine should be used as soon as practicable. If storage is necessary, hold at<br />

+2°C to +8°C for not more than 24 hours.<br />

Infanrix hexa must be reconstituted by adding the entire contents of the syringe<br />

to the vial and shaking until the pellet is completely dissolved. Reconstituted<br />

vaccine should be used as soon as practicable. If storage is necessary, hold at<br />

room temperature for not more than 8 hours.<br />

Menitorix must be reconstituted by adding the entire contents of the diluent<br />

syringe to the vial and shaking until the powder is completely dissolved.<br />

Reconstituted vaccine should be used as soon as practicable. If storage is<br />

necessary, hold at +2°C to +8°C for not more than 24 hours.<br />

4.3.6 Dosage and administration<br />

<strong>The</strong> dose of all Hib-containing vaccines is 0.5 mL to be given by IM injection.<br />

Co-administration with other vaccines<br />

All Hib-containing vaccines may be administered in separate sites on the same<br />

day as any of the other childhood vaccines such as pneumococcal conjugate,<br />

meningococcal serogroup C conjugate (MenCCV), hepatitis B, DTPa-containing<br />

and inactivated poliomyelitis vaccine (IPV) (or IPV-containing) vaccines.<br />

No or minimal immunologic interference has been observed when children are<br />

vaccinated with pneumococcal conjugate vaccines (7vPCV – Prevenar; 13vPCV<br />

– Prevenar 13; or 10vPCV – Synflorix) and PRP-T-containing hexavalent vaccine<br />

(Infanrix hexa) at the same immunisation visit. 21-24<br />

Interchangeability of Hib vaccines<br />

Where possible, the same brand of Hib-containing vaccine should be used for<br />

all primary doses. If different Hib-containing vaccines (i.e. PRP-OMP and PRP-T<br />

vaccines) are used in the primary series, then 3 doses (of any Hib-containing<br />

vaccine) are required at 2, 4 and 6 months of age, with a booster of a Hibcontaining<br />

vaccine at 12 months of age. For booster doses and in children<br />

>15 months of age, regardless of previous Hib vaccinations, a single dose of any<br />

Hib-containing vaccine is sufficient for protection.<br />

194 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.3.7 Recommendations<br />

Infants<br />

A Hib-containing vaccine is recommended for all infants from 2 months of age.<br />

PRP-T-containing Hib vaccines require 3 primary doses, at 2, 4 and 6 months of<br />

age, followed by a booster dose at 12 months of age. <strong>Immunisation</strong> using PRP-<br />

OMP-containing Hib vaccine only requires 2 primary doses, at ages 2 and<br />

4 months, followed by a booster at 12 months of age.<br />

<strong>The</strong> 1st dose of a Hib-containing vaccine can be given as early as 6 weeks of age.<br />

If the 1st dose is given at 6 weeks of age, the next scheduled doses should still be<br />

given at 4 months and 6 months of age.<br />

Booster doses<br />

A single booster dose of Hib vaccine is recommended at 12 months of age<br />

(see ‘Infants’ above). This booster dose can be administered using either the<br />

monovalent Hib vaccine or, where meningococcal serogroup C vaccination is<br />

also scheduled, the combined Hib-meningococcal serogroup C conjugate vaccine<br />

(Hib-MenCCV).<br />

Children aged ≥15 months and up to 59 months of age at presentation who<br />

have not received a primary course of a Hib or Hib-containing vaccine will only<br />

require 1 dose of vaccine as catch-up, irrespective of the number of previous<br />

doses administered. <strong>The</strong>re should be a minimum 2-month interval between their<br />

last dose and the catch-up dose. Catch-up for Hib vaccination for children up to<br />

59 months of age is outlined in Table 2.1.8 Catch-up schedule for Hib vaccination for<br />

children


according to the recommendations above and Table 2.1.8 Catch-up schedule for<br />

Hib vaccination for children


4.3.11 Public health management of invasive Hib disease<br />

Haemophilus influenzae type b is a notifiable disease in all states and territories<br />

in Australia.<br />

Further instructions about the public health management of Hib, including<br />

management of cases of invasive Hib disease and their contacts, should<br />

be obtained from state/territory public health authorities (see Appendix 1<br />

Contact details for <strong>Australian</strong>, state and territory government health authorities and<br />

communicable disease control).<br />

4.3.12 Variations from product information<br />

<strong>The</strong> product information for Infanrix hexa states that this vaccine is indicated<br />

for primary immunisation of infants from the age of 6 weeks. <strong>The</strong> ATAGI<br />

recommends that this vaccine may also be used for catch-up of the primary<br />

schedule in children


4.4 HEPATITIS A<br />

4.4.1 Virology<br />

Hepatitis A is an acute infection of the liver caused by the hepatitis A virus<br />

(HAV), a picornavirus (a small single-stranded RNA virus). 1 <strong>The</strong> virus survives<br />

well in the environment outside of the human host. It persists on hands for<br />

several hours and in food kept at room temperature for considerably longer, and<br />

is relatively resistant to heat and freezing.<br />

4.4.2 Clinical features<br />

Hepatitis A is an infection of humans; there is no animal reservoir. 1 HAV is<br />

predominantly transmitted by the faecal–oral route. <strong>The</strong> infecting dose is<br />

unknown, but it is presumed to be low. <strong>The</strong> incubation period of hepatitis A is 15<br />

to 50 days, with a mean of about 28 days. 2 HAV is excreted in faeces for up to 2<br />

weeks before the onset of illness and for at least 1 week afterwards. 1<br />

In young children, HAV usually causes either an asymptomatic infection or a<br />

very mild illness without jaundice; adults are more likely to have symptomatic<br />

infection (over 70%). 2 Patients with symptomatic illness typically have a 4- to<br />

10-day prodrome of systemic (fever, malaise, weakness and anorexia) and<br />

gastrointestinal (nausea and vomiting) symptoms. Dark urine is usually the first<br />

specific manifestation of acute hepatitis A infection, followed a day or two later<br />

by jaundice and pale faeces. 2 <strong>The</strong> prodromal symptoms tend to wane with the<br />

onset of jaundice, although the anorexia and malaise may persist; pruritus and<br />

localised hepatic discomfort or pain may follow. 1 <strong>The</strong> duration of illness varies,<br />

but most patients feel better and have normal, or near normal, liver function<br />

tests within a month of the onset of illness. 3 Complications of hepatitis A are<br />

uncommon but include, on rare occasions, fulminant hepatitis. 4 <strong>The</strong> case-fatality<br />

rate of hepatitis A increases with age. 2 Hepatitis A does not cause chronic liver<br />

disease. Relapse has been found in up to 10% of cases, but recovery is universal.<br />

HAV does not cause chronic infection and immunity after infection is life-long. 2<br />

Diagnosis of hepatitis A is made by detecting anti-HAV IgM in serum during the<br />

acute illness. Anti-HAV IgM is invariably present by the time the patient presents<br />

and persists for 3 to 6 months after the acute illness. 1 Serum anti-HAV IgG alone<br />

indicates past infection (or possibly immunisation) and therefore immunity; it<br />

probably persists for life. 1<br />

4.4.3 Epidemiology<br />

Hepatitis A was a considerable public health problem in Australia in the 1990s.<br />

During this time, numerous outbreaks occurred in child day-care centres and<br />

preschools, 5 Indigenous communities, 6 communities of men who have sex with<br />

men, 7 schools and residential facilities for the disabled, 8 and communities of<br />

persons who inject drugs. 7 A very large outbreak of hepatitis A, associated with<br />

the consumption of raw oysters, occurred in New South Wales in 1997 and there<br />

was a large outbreak associated with semidried tomatoes during 2009. 9,10<br />

198 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


In recent years, hepatitis A notifications and hospitalisations have been low with<br />

a downward trend. 11 This has been accompanied by an increasing proportion of<br />

cases related to travel to countries where hepatitis A is endemic. 12-14 Advocacy<br />

for hepatitis A vaccination of travellers and those at increased risk because of<br />

lifestyle or occupation remains a priority, as does the hepatitis A vaccination<br />

program for Aboriginal and Torres Strait Islander children. Established<br />

initially in north Queensland in 1999 for Indigenous children aged 18 months, 6<br />

the hepatitis A vaccination program was expanded in 2005 to include all<br />

Indigenous children aged ≤2 years in the Northern Territory, Queensland, South<br />

Australia and Western Australia, contributing substantially to the decline in<br />

notifications. 15,16 In north Queensland, most Indigenous children >2 years of<br />

age have now been immunised against hepatitis A. However, it is important to<br />

note that Indigenous children remain at considerably greater risk – not only of<br />

acquiring hepatitis A, but also for being hospitalised with the infection – than<br />

non-Indigenous children. 11,17 This is particularly true for Indigenous children<br />

residing in other regions of Queensland, the Northern Territory, South Australia<br />

and Western Australia. (See also 3.1 Vaccination for Aboriginal and Torres Strait<br />

Islander people.)<br />

4.4.4 Vaccines<br />

Monovalent hepatitis A vaccines<br />

• Avaxim – Sanofi Pasteur Pty Ltd (formaldehyde-inactivated hepatitis<br />

A virus [GBM strain]). Each 0.5 mL pre-filled syringe contains 160<br />

antigen units of hepatitis A virus (HAV) antigens inactivated by<br />

formaldehyde; 0.3 mg aluminium as aluminium hydroxide; 2.5 µL<br />

phenoxyethanol; 12.5 µg formaldehyde; traces of neomycin and<br />

bovine serum albumin.<br />

• Havrix Junior – GlaxoSmithKline (formaldehyde-inactivated<br />

hepatitis A virus [HM175 strain]). Each 0.5 mL monodose vial or<br />

pre-filled syringe contains 720 ELISA units of HAV antigens; 0.25 mg<br />

aluminium as aluminium hydroxide; traces of formaldehyde,<br />

neomycin and polysorbate 20.<br />

• Havrix 1440 – GlaxoSmithKline (formaldehyde-inactivated<br />

hepatitis A virus [HM175 strain]). Each 1.0 mL monodose vial or<br />

pre-filled syringe contains 1440 ELISA units of HAV antigens; 0.5<br />

mg aluminium as aluminium hydroxide; traces of formaldehyde,<br />

neomycin and polysorbate 20.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 199<br />

4.4 HEPATITIS A


• Vaqta Paediatric/Adolescent formulation – CSL Limited/Merck & Co<br />

Inc (formaldehyde-inactivated hepatitis A virus [CR326F strain]). Each<br />

0.5 mL monodose vial or pre-filled syringe contains approximately<br />

25 units (U) of hepatitis A virus protein; 0.225 mg aluminium as<br />

aluminium hydroxide; 35 µg borax; traces of formaldehyde, neomycin<br />

and bovine serum albumin.<br />

• Vaqta Adult formulation – CSL Limited/Merck & Co Inc<br />

(formaldehyde-inactivated hepatitis A virus [CR326F strain]). Each<br />

1.0 mL monodose vial or pre-filled syringe contains approximately<br />

50 U of hepatitis A virus protein; 0.45 mg aluminium as aluminium<br />

hydroxide; 70 µg borax; traces of formaldehyde, neomycin and bovine<br />

serum albumin.<br />

Combination vaccines that contain hepatitis A<br />

• Twinrix Junior (360/10) – GlaxoSmithKline (formaldehyde-inactivated<br />

hepatitis A virus [HM175 strain] and recombinant hepatitis B vaccine).<br />

Each 0.5 mL monodose vial or pre-filled syringe contains 360 ELISA<br />

units of HAV antigens, 10 µg recombinant DNA hepatitis B surface<br />

antigen protein; 0.225 mg aluminium as aluminium phosphate/<br />

hydroxide; traces of formaldehyde, neomycin, trometamol and<br />

polysorbate 20. May contain yeast proteins.<br />

• Twinrix (720/20) – GlaxoSmithKline (formaldehyde-inactivated<br />

hepatitis A virus [HM175 strain] and recombinant hepatitis B vaccine).<br />

Each 1.0 mL monodose vial or pre-filled syringe contains 720 ELISA<br />

units of HAV antigens, 20 µg recombinant DNA hepatitis B surface<br />

antigen protein; 0.45 mg aluminium as aluminium phosphate/<br />

hydroxide; traces of formaldehyde, neomycin, trometamol and<br />

polysorbate 20. May contain yeast proteins.<br />

• Vivaxim – Sanofi Pasteur Pty Ltd (formaldehyde-inactivated hepatitis A<br />

virus [GBM strain] and typhoid Vi capsular polysaccharide). Supplied<br />

in a dual-chamber syringe which enables the two vaccines to be mixed<br />

just before administration. Each 1.0 mL dose of mixed vaccine contains<br />

160 ELISA units of inactivated hepatitis A virus antigens, 25 µg purified<br />

typhoid Vi capsular polysaccharide strain Ty2; 0.3 mg aluminium as<br />

aluminium hydroxide; 2.5 µL phenoxyethanol; 12.5 µg formaldehyde;<br />

traces of neomycin, bovine serum albumin and polysorbate 80.<br />

200 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Inactivated hepatitis A vaccines are prepared from HAV harvested from<br />

human diploid cell cultures, which are then purified by ultrafiltration and<br />

chromatography, inactivated by formaldehyde, and adsorbed onto aluminium<br />

hydroxide adjuvant. Although the vaccines are prepared from differing strains of<br />

HAV, there is only one known serotype; immunity induced by a particular strain<br />

probably provides protection against all strains. 1<br />

Inactivated hepatitis A vaccines induce HAV antibodies (anti-HAV) at titres<br />

many-fold greater than are provided by the recommended dose of normal<br />

human immunoglobulin. Although the vaccines are highly immunogenic (see<br />

below), antibody titres are usually below the detection limits of the routinely<br />

available commercial tests for anti-HAV. 1 <strong>The</strong>refore, serological testing to assess<br />

immunity after vaccination against hepatitis A is neither necessary nor appropriate.<br />

Likewise, it is also inappropriate to undertake testing if an individual cannot<br />

recall if he/she has been vaccinated against hepatitis A in the past; if no<br />

vaccination records are available, vaccination should be advised. However,<br />

certain groups of people should be screened for natural immunity to hepatitis A<br />

to avoid unnecessary vaccination: those born before 1950; those who spent their<br />

early childhood in endemic areas; and those with an unexplained previous<br />

episode of hepatitis or jaundice. In addition, it is necessary to test for other causes<br />

of hepatitis, in particular hepatitis B, in those with unexplained jaundice.<br />

Hepatitis A vaccines are highly immunogenic in both children and adults,<br />

with virtually universal seroconversion 4 weeks after vaccination. 1,18,19 Two<br />

randomised clinical trials conducted in the early 1990s showed that the<br />

vaccines have a very high protective efficacy, approaching 100%. 20,21 This<br />

finding is supported by the apparent eradication of hepatitis A from Indigenous<br />

communities in north Queensland and the Northern Territory since the<br />

introduction of the vaccination program in those regions. 6,16<br />

<strong>The</strong> duration of immunity, and therefore protection, following vaccination is<br />

not certain. However, vaccine-induced anti-HAV probably persists for many<br />

years. <strong>The</strong>re is no current evidence that booster doses are required; in healthy<br />

individuals, it is quite possible that they will never be required. 22<br />

4.4.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 23<br />

Store at +2°C to +8°C. Do not freeze.<br />

4.4.6 Dosage and administration<br />

Inactivated hepatitis A vaccines are to be given by IM injection.<br />

<strong>The</strong> recommended doses and schedules are shown in Table 4.4.1.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 201<br />

4.4 HEPATITIS A


Table 4.4.1: Recommended doses and schedules for use of inactivated hepatitis<br />

A and hepatitis A combination vaccines*<br />

Vaccine<br />

Age of<br />

vaccine<br />

recipient<br />

(years)<br />

Dose<br />

(HAV<br />

antigen)<br />

Monovalent hepatitis A vaccines<br />

Volume<br />

per dose<br />

(mL)<br />

Number<br />

of doses<br />

202 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Vaccination<br />

schedule<br />

Avaxim ≥2 160 ELISA U 0.5 2 1st dose: day 0 (day of vaccination)<br />

2nd dose: 6 to 12 months after 1st dose<br />

Havrix<br />

Junior<br />

Havrix<br />

1440<br />

Vaqta<br />

Paediatric/<br />

Adolescent<br />

Vaqta<br />

Adult<br />

2–


Co-administration with other vaccines<br />

Hepatitis A vaccines are inactivated vaccines and can be administered either<br />

simultaneously with, or at any time before or after, all other vaccines relevant<br />

to international travel. 24<br />

Combination hepatitis A/hepatitis B vaccines can be administered<br />

simultaneously with, or at any time before or after, all other vaccines relevant<br />

to international travel.<br />

<strong>The</strong> combination hepatitis A/typhoid vaccine can be administered<br />

simultaneously with, or at any time before or after, all other vaccines relevant<br />

to international travel.<br />

Interchangeability of hepatitis A vaccines<br />

Although the manufacturers use slightly different production methods and<br />

quantify the HAV antigen content in their respective vaccines differently, the<br />

hepatitis A vaccines of the different manufacturers used in ‘equivalent’ schedules<br />

in Table 4.4.1 can be considered interchangeable, when given in a 2-dose course.<br />

As there is only one brand of combination hepatitis A/hepatitis B vaccine,<br />

interchangeability is not relevant. (See also ‘Recommendations for the use of<br />

combination hepatitis A/hepatitis B vaccines’ in 4.4.7 Recommendations below.)<br />

4.4.7 Recommendations<br />

Hepatitis A vaccination is recommended for persons with an increased risk<br />

of acquiring hepatitis A and/or who are at increased risk of severe disease.<br />

Serological testing for immunity to hepatitis A from previous infection is<br />

not usually required prior to vaccination, but may be indicated in some<br />

circumstances (see ‘Serological testing for hepatitis A immunity from infection<br />

and/or vaccination’ below).<br />

When vaccination against both hepatitis A and hepatitis B (or hepatitis A and<br />

typhoid) is indicated, combination vaccines may be used, as described below.<br />

Recommendations for hepatitis A vaccine<br />

Hepatitis A vaccination is recommended for the following groups:<br />

Aboriginal and Torres Strait Islander children residing in the Northern Territory, Queensland,<br />

South Australia and Western Australia<br />

Two doses of hepatitis A vaccine are required for Aboriginal and Torres Strait<br />

Islander children living in these jurisdictions, due to the increased risk for<br />

hepatitis A in this population (see 4.4.3 Epidemiology above). Vaccination for<br />

these children should commence in the 2nd year of life, with the 1st dose given<br />

between 12 and 18 months of age, and the 2nd dose given between 18 and 24<br />

months of age. <strong>The</strong> recommended interval between doses is 6 months (see<br />

Table 4.4.1). State/territory health authorities should be contacted about local<br />

hepatitis A vaccination schedules, including catch-up.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 203<br />

4.4 HEPATITIS A


Travellers (≥1 year of age) to hepatitis A endemic areas<br />

Travellers to (≥1 year of age), and expatriates living in, moderately to highly<br />

endemic areas for hepatitis A should receive hepatitis A vaccine. 25 A single dose<br />

of a monovalent hepatitis A vaccine provides protective levels of anti-HAV for at<br />

least a year; 1 a 2nd dose is recommended 6 to 12 months following the 1st dose,<br />

to increase the duration of protection (see Table 4.4.1).<br />

Persons whose occupation puts them at increased risk of acquiring hepatitis A<br />

Persons whose occupation puts them at increased risk of acquiring hepatitis A<br />

include: persons who live or work in rural and remote Indigenous communities<br />

and/or persons who regularly provide care for Aboriginal and Torres Strait<br />

Islander children in the Northern Territory, Queensland, South Australia and<br />

Western Australia; staff working in early childhood education and care; carers<br />

of persons with developmental disabilities; and plumbers or sewage workers.<br />

See also 3.3 Groups with special vaccination requirements, Table 3.3.7 Recommended<br />

vaccinations for persons at increased risk of certain occupationally acquired vaccinepreventable<br />

diseases.<br />

Persons whose lifestyle puts them at increased risk of acquiring hepatitis A<br />

Persons who engage in anal intercourse, men who have sex with men, persons<br />

who inject drugs (including inmates of correctional facilities) and sex industry<br />

workers are at increased risk of acquiring hepatitis A. 26 See also 3.3 Groups with<br />

special vaccination requirements.<br />

Persons with developmental disabilities<br />

Vaccination is recommended for persons with developmental disabilities, and<br />

susceptible carers, who attend both residential and non-residential facilities for<br />

persons with developmental disabilities. Although conditions/measures to limit<br />

the likelihood of hepatitis A transmission in such facilities have improved in<br />

recent decades, outbreaks of hepatitis A can occur in these settings. 26<br />

Persons with chronic liver disease, liver solid organ transplant recipients and/or those chronically<br />

infected with either hepatitis B or hepatitis C viruses<br />

Hepatitis A vaccination is recommended for persons with chronic liver disease<br />

of any aetiology. 2,26 Those with chronic liver disease of mild to moderate severity<br />

mount a satisfactory immune response following vaccination, but those with<br />

end-stage liver disease do not respond as well, and liver transplant recipients<br />

may not respond at all. 27,28 Nevertheless, all those with chronic liver disease who<br />

are non-immune to hepatitis A should be vaccinated, preferably as early in the<br />

course of the disease as possible.<br />

Vaccination is recommended for persons with chronic hepatitis C and hepatitis B<br />

infection because of the high case-fatality rate among these persons if they<br />

acquire hepatitis A. 2<br />

204 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Recommendations for the use of combination hepatitis A/hepatitis B vaccines<br />

Combination hepatitis A/hepatitis B vaccines should be considered for<br />

susceptible persons in whom both hepatitis A and hepatitis B vaccines are<br />

recommended. Vaccination is usually provided in a 3-dose schedule (see Table<br />

4.4.1). Twinrix (720/20) can be administered in a 2-dose regimen in persons 1 to<br />


fever. This combination is particularly useful for those already immunised<br />

against hepatitis B.<br />

To provide longer-term protection against hepatitis A, a single dose of a<br />

monovalent adult formulation hepatitis A vaccine administered between 6 and<br />

36 months after the single dose of combination hepatitis A/typhoid vaccine<br />

is required (see Table 4.4.1). If there is a continued risk of typhoid infection, a<br />

booster dose of parenteral typhoid Vi polysaccharide vaccine is required<br />

3 years after the single dose of combination hepatitis A/typhoid vaccine. <strong>The</strong><br />

combination hepatitis A/typhoid vaccine may be used as a ‘booster’ vaccine<br />

for hepatitis A if a person received a previous dose of a monovalent adult<br />

formulation hepatitis A vaccine; this should be given at a minimum interval of<br />

6 months after the 1st dose of hepatitis A vaccine.<br />

Serological testing for hepatitis A immunity from infection and/or vaccination<br />

Serological testing for immunity to hepatitis A is not recommended before<br />

routine administration of hepatitis A vaccine to those in most of the categories<br />

above, for example, Aboriginal and Torres Strait Islander children or travellers.<br />

However, previous infection with hepatitis A is more likely to have occurred in<br />

persons born before 1950, those who spent their early childhood in an endemic<br />

area, and those with an unexplained previous episode of hepatitis or jaundice.<br />

In such persons, testing for total hepatitis A antibodies or anti-HAV IgG may be<br />

indicated, and, if positive, indicates immunity to hepatitis A. Such persons do not<br />

need hepatitis A vaccination.<br />

Interpretation of the results of serological testing may be enhanced by discussion<br />

with the laboratory that performed the test, ensuring that relevant clinical<br />

information is provided.<br />

Serological testing following vaccination is not routinely required.<br />

4.4.8 Pregnancy and breastfeeding<br />

Hepatitis A vaccine is not routinely recommended for pregnant or breastfeeding<br />

women, but can be given where vaccination is considered necessary (see 4.4.7<br />

Recommendations above).<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.4.9 Contraindications<br />

<strong>The</strong> only absolute contraindications to hepatitis A vaccines are:<br />

• anaphylaxis following a previous dose of any hepatitis A vaccine<br />

• anaphylaxis following any vaccine component.<br />

Combination vaccines containing the hepatitis B component are contraindicated<br />

in persons with a history of anaphylaxis to yeast.<br />

206 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.4.10 Adverse events<br />

<strong>The</strong> most common adverse events following administration of hepatitis A<br />

vaccines are mild local events of a short duration, probably caused by the<br />

aluminium hydroxide adjuvant. About 15% of adults report headache and<br />

approximately 5% report malaise or fatigue following vaccination. 26 Up to 20% of<br />

children who receive either Havrix or Vaqta experience soreness at the injection<br />

site. In both adults and children, systemic adverse events such as headache and<br />

fever are much less common than local adverse events. 26<br />

Hepatitis A vaccines do not affect liver enzyme levels. <strong>The</strong>y can be safely given<br />

to persons with HIV infection, and do not adversely affect either the HIV load or<br />

CD4 + cell count. 30<br />

4.4.11 Public health management of hepatitis A<br />

Hepatitis A is a notifiable disease in all states and territories in Australia. Detailed<br />

information regarding the management of hepatitis A cases and contacts can be<br />

found in the national guidelines for control of hepatitis A31 (www.health.gov.au/<br />

cdnasongs).<br />

Further instructions can also be obtained from state/territory public health<br />

authorities (see Appendix 1 Contact details for <strong>Australian</strong>, state and territory<br />

government health authorities and communicable disease control).<br />

Post-exposure prophylaxis using hepatitis A vaccine or normal human<br />

immunoglobulin (NHIG) can be used to prevent secondary cases in close<br />

contacts of hepatitis A cases. However, vaccination is recommended in preference<br />

to NHIG for use in post-exposure prophylaxis in persons ≥12 months of age who<br />

are immunocompetent (see Part 5 Passive immunisation). 31<br />

4.4.12 Variations from product information<br />

None.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

PART 4 VACCINE-PREVENTABLE DISEASES 207<br />

4.4 HEPATITIS A


4.5 HEPATITIS B<br />

4.5.1 Virology<br />

Hepatitis B virus (HBV) contains circular, partially double-stranded DNA. <strong>The</strong><br />

outer surface of the virus is glycolipid, which contains the hepatitis B surface<br />

antigen (HBsAg). Other important antigenic components are the hepatitis B core<br />

antigen (HBcAg) and hepatitis B e antigen (HBeAg). HBcAg is not detectable<br />

in serum, but can be detected in liver tissue in persons with acute or chronic<br />

hepatitis B infection. HBeAg, and antibodies against HBeAg (anti-HBe) or the<br />

HBcAg (anti-HBc), are serological markers of HBV infection. Antibodies against<br />

HBsAg (anti-HBs) indicate immunity, which may result from either natural<br />

infection or immunisation (in which case there would not be any markers of HBV<br />

infection). Persistence of HBsAg denotes infectivity, which is greater if HBeAg<br />

and/or HBV DNA are also positive. 1 Occult hepatitis B infection is characterised<br />

by the presence of HBV DNA in the liver (with or without detectable HBV DNA<br />

in the serum) and negative HBsAg. 2<br />

4.5.2 Clinical features<br />

In approximately 30 to 50% of adults, infection causes symptomatic acute<br />

hepatitis, but in neonates and young children, particularly those


4.5.3 Epidemiology<br />

<strong>The</strong> prevalence of chronic HBV infection differs in different parts of the world,<br />

and may be quite variable within countries. <strong>The</strong> prevalence of chronic HBV<br />

infection varies from less than 0.5% among Caucasians in the United States,<br />

northern Europe and Australia, 1 to 5% in the Mediterranean countries, parts<br />

of eastern Europe, Africa, Central and South America, up to greater than 10%<br />

in many sub-Saharan African, East and Southeast Asian and Pacific island<br />

populations. 7-10 In regions of moderate to high prevalence of HBsAg (where ≥2%<br />

of the population is HBsAg-positive), infections are mainly acquired perinatally<br />

or in early childhood. 1<br />

Chronic infection and its sequelae, including cirrhosis and hepatocellular<br />

carcinoma, contribute to the majority of HBV disease burden in Australia. In<br />

recent decades, the burden from such disease has been increasing, concurrent<br />

with the increasing number of immigrants from regions of high HBV<br />

prevalence. 11 Aboriginal and Torres Strait Islander people, and migrants born<br />

in Asia and Pacific islands, North Africa, Middle Eastern and Mediterranean<br />

countries, have a significantly increased prevalence of chronic HBV infection<br />

compared with the rest of the <strong>Australian</strong>-born population. 12,13 First-generation<br />

immigrants of culturally and linguistically diverse background, who are<br />

mostly from countries of high HBV endemicity, usually retain the prevalence<br />

of chronic HBV infection of their country of origin. Other population groups<br />

with an increased prevalence of markers of HBV infection include patients with<br />

HIV infection, persons who used injected drugs between 1980 and 1990, and<br />

household contacts of someone diagnosed with hepatitis between 1980 and<br />

1990. 13 Notification of chronic HBV infection depends on levels of hepatitis B<br />

testing and reporting, and a substantial proportion of persons with chronic HBV<br />

infection remain undiagnosed. It has been estimated by mathematical modelling<br />

that, in 2010, about 170 000 people were living with HBV infection in Australia,<br />

with about 335 deaths due to HBV infection in that year. 14<br />

Newly acquired cases of HBV infection in Australia mostly occur in young<br />

adults, through injecting drug use, skin penetration procedures or sexual<br />

contact. 15 Between 2006 and 2010, the notification rate of newly acquired<br />

hepatitis B in Australia ranged from 1.0 to 1.4 per 100 000 population.<br />

Since 2001, the rate of diagnosis of newly acquired infections has declined<br />

substantially among people aged 15–29 years and has remained relatively stable<br />

among people aged ≥30 years. 14-16 However, some new HBV infections are<br />

asymptomatic and may go undetected.<br />

Similar to chronic infection, higher rates of notified cases of newly acquired<br />

hepatitis B, or hospitalisation due to acute hepatitis B, have been reported<br />

among Aboriginal and Torres Strait Islander people compared with the general<br />

<strong>Australian</strong> population. 17,18 In one United States study, adults with diabetes<br />

mellitus had a greater chance of developing acute hepatitis B disease than<br />

PART 4 VACCINE-PREVENTABLE DISEASES 209<br />

4.5 HEPATITIS B


the general population; 19 however, there are no published <strong>Australian</strong> studies<br />

examining this.<br />

Transmission of HBV may result from inoculation through broken or penetrated<br />

skin, or by mucosal contact with blood or other body fluids (mainly vaginal<br />

fluids and semen) from an infectious person. <strong>The</strong>re are four major routes of<br />

HBV transmission:<br />

• perinatal transmission from infected mother to neonate (vertical<br />

transmission), usually occurring at or around the time of birth<br />

• parenteral or mucosal exposure to infected blood and other bodily fluids;<br />

common scenarios include:<br />

» sharing of contaminated equipment that penetrates the skin, such as<br />

needles (among persons who inject drugs), tattoo equipment, bodypiercing<br />

equipment, acupuncture equipment and razor blades<br />

» needle-stick injury, for example, in a healthcare setting<br />

» contact between infective body fluids and mucous membranes<br />

• sexual contact (including vaginal or anal intercourse, although the latter is<br />

associated with a higher risk)<br />

• non-sexual contact with an infected person (horizontal transmission),<br />

including household transmission, for example, child-to-child transmission<br />

through contact between open sores or wounds.<br />

In Australia, screening of blood and organ donors using nucleic acid<br />

amplification testing has virtually eliminated the risk of transmission of<br />

hepatitis B through blood transfusion and organ transplants. 20,21 Saliva may<br />

contain levels of virus that are likely to be infective only if inoculated directly<br />

into tissue (ocular or mucous membranes). <strong>The</strong> risk of transmission by<br />

inadvertent inoculation by other means, such as by toothbrush, razor etc., or<br />

through close personal contact in households in which one or more infected<br />

persons reside, is low but not negligible. 22-29<br />

<strong>The</strong> strategy for prevention of hepatitis B through immunisation in Australia<br />

commenced in the early 1980s, with vaccination programs targeting individuals<br />

with increased risk of HBV exposure, including infants at particular<br />

risk of infection at birth. Universal infant vaccination commenced in the<br />

Northern Territory in 1990. A universal hepatitis B vaccination program was<br />

recommended for infants and adolescents in 1996. <strong>The</strong> adolescent program<br />

commenced in some states and territories in 1997 and the universal infant<br />

program, which includes a dose given at birth, began nationally in 2000. <strong>The</strong><br />

adolescent program will continue until those immunised for hepatitis B in the<br />

infant program reach adolescence.<br />

210 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.5.4 Vaccines<br />

Monovalent hepatitis B vaccines<br />

• Engerix-B – GlaxoSmithKline (recombinant DNA hepatitis B vaccine).<br />

Adult formulation – Each 1.0 mL monodose vial or pre-filled syringe<br />

contains 20 µg recombinant hepatitis B surface antigen (HBsAg)<br />

protein, adsorbed onto 0.5 mg aluminium as aluminium hydroxide.<br />

Paediatric formulation – Each 0.5 mL monodose vial or pre-filled<br />

syringe contains 10 µg HBsAg protein, adsorbed onto 0.25 mg<br />

aluminium as aluminium hydroxide.<br />

Both formulations may contain yeast proteins.<br />

• H-B-Vax II – CSL Limited/Merck & Co Inc (recombinant DNA<br />

hepatitis B vaccine). Adult formulation – Each 1.0 mL monodose<br />

vial or pre-filled syringe contains 10 µg recombinant HBsAg protein,<br />

adsorbed onto 0.5 mg aluminium hydroxide. Paediatric formulation<br />

– Each 0.5 mL monodose vial or pre-filled syringe contains 5 µg<br />

recombinant HBsAg protein, adsorbed onto 0.25 mg aluminium<br />

hydroxide. Dialysis formulation – Each 1.0 mL monodose vial<br />

contains 40 µg recombinant HBsAg protein, adsorbed onto 0.5 mg<br />

aluminium hydroxide.<br />

All formulations may contain yeast proteins.<br />

Combination vaccines that contain hepatitis B<br />

• Infanrix hexa – GlaxoSmithKline (DTPa-hepB-IPV-Hib; diphtheriatetanus-acellular<br />

pertussis-hepatitis B-inactivated poliovirus-<br />

Haemophilus influenzae type b). <strong>The</strong> vaccine consists of both a 0.5 mL<br />

pre-filled syringe containing ≥30 IU diphtheria toxoid, ≥40 IU tetanus<br />

toxoid, 25 µg pertussis toxoid, 25 µg filamentous haemagglutinin, 8 µg<br />

pertactin, 10 µg recombinant HBsAg, 40 D-antigen units inactivated<br />

poliovirus type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1)<br />

and 32 D-antigen units type 3 (Saukett), adsorbed onto aluminium<br />

hydroxide/phosphate; traces of formaldehyde, polysorbate 80,<br />

polysorbate 20, polymyxin and neomycin; and a vial containing a<br />

lyophilised pellet of 10 µg purified Hib capsular polysaccharide (PRP)<br />

conjugated to 20–40 µg tetanus toxoid. May contain yeast proteins.<br />

• Twinrix Junior (360/10) – GlaxoSmithKline (formaldehyde-inactivated<br />

hepatitis A virus [HM175 strain] and recombinant hepatitis B vaccine).<br />

Each 0.5 mL monodose vial or pre-filled syringe contains 360 ELISA<br />

units of HAV antigens, 10 µg recombinant DNA hepatitis B surface<br />

antigen protein; 0.225 mg aluminium as aluminium phosphate/<br />

PART 4 VACCINE-PREVENTABLE DISEASES 211<br />

4.5 HEPATITIS B


hydroxide; traces of formaldehyde, neomycin, trometamol and<br />

polysorbate 20. May contain yeast proteins.<br />

• Twinrix (720/20) – GlaxoSmithKline (formaldehyde-inactivated<br />

hepatitis A virus [HM175 strain] and recombinant hepatitis B vaccine).<br />

Each 1.0 mL monodose vial or pre-filled syringe contains 720 ELISA<br />

units of HAV antigens, 20 µg recombinant DNA hepatitis B surface<br />

antigen protein; 0.45 mg aluminium as aluminium phosphate/<br />

hydroxide; traces of formaldehyde, neomycin, trometamol and<br />

polysorbate 20. May contain yeast proteins.<br />

Hepatitis B vaccines are prepared using recombinant technology. After<br />

purification, the HBsAg protein is adsorbed onto elemental aluminium (as<br />

hydroxide and/or phosphate). Hepatitis B vaccines may contain up to 1% yeast<br />

proteins (but no yeast DNA).<br />

<strong>The</strong> Engerix-B and the H-B-Vax II vaccines are manufactured by different<br />

processes, and the HBsAg content of an ‘equivalent’ dose of these 2 vaccines is<br />

different. Studies of hepatitis B vaccines have been conducted using different<br />

schedules and intervals for different age groups. Acceptable schedules are shown<br />

in Table 4.5.1 and are described below.<br />

<strong>The</strong> standard 3-dose schedule and variations<br />

Neonates, children and young adults aged


For older children and young adults aged


of seroconversion increases progressively from approximately 35% after the 1st<br />

dose to more than 90% after the 3rd dose. <strong>The</strong>re is evidence of immunity in most<br />

vaccine recipients after administration of 2 doses of a 3-dose schedule. However,<br />

the 3rd dose is necessary to increase the percentage of responders and to provide<br />

long-term protection.<br />

Alternative 2-dose schedule for adolescents<br />

Several studies have demonstrated that adolescents 11–15 years of age who<br />

receive 2 doses of adult formulation monovalent hepatitis B vaccine 4 to<br />

6 months apart develop similar protective antibody levels to those vaccinated<br />

using paediatric formulations in the standard 3-dose schedule. 41-43<br />

Using a 2-dose schedule for the 11–15 years age group may improve<br />

compliance and will provide comparable immunogenicity to that of a 3-dose<br />

paediatric schedule. Adolescents (11–15 years of age) can be vaccinated with<br />

the adult formulation of either H-B-Vax II or Engerix-B in a 2-dose schedule<br />

(see Table 4.5.1).<br />

Table 4.5.1: Recommended schedules for use of monovalent hepatitis B and<br />

hepatitis B combination vaccines<br />

Vaccine<br />

Age of<br />

vaccine<br />

recipient<br />

Recommended infant schedule<br />

Engerix-B (paediatric<br />

formulation)<br />

or<br />

H-B-Vax II (paediatric<br />

formulation)<br />

Combination hepatitis<br />

B-containing vaccine<br />

(e.g. Infanrix hexa<br />

(DTPa-hepB-IPV-Hib)<br />

birth<br />

2, 4 and 6 ‡<br />

months<br />

Dose<br />

(HBsAg<br />

protein)<br />

10 µg<br />

(Engerix-B)<br />

or<br />

5 µg<br />

(H-B-Vax II)<br />

Volume per<br />

dose (mL)<br />

Number of<br />

doses<br />

0.5 1<br />

10 µg 0.5 3<br />

Monovalent hepatitis B vaccines – standard 3-dose schedule<br />

Engerix-B (paediatric<br />

formulation)<br />

Engerix-B (adult<br />

formulation)<br />

H-B-Vax II (paediatric<br />

formulation)<br />


Vaccine<br />

Age of<br />

vaccine<br />

recipient<br />

Dose<br />

(HBsAg<br />

protein)<br />

Volume per<br />

dose (mL)<br />

Number of<br />

doses<br />

Monovalent hepatitis B vaccines – standard 3-dose schedule<br />

H-B-Vax II (adult<br />

formulation)<br />

H-B-Vax II (dialysis<br />

formulation)<br />

≥20 years 10 µg 1.0 3<br />

≥20 years 40 µg 1.0 3<br />

Recommended schedule<br />

intervals* †<br />

1st dose: day 0 (day of<br />

vaccination)<br />

2nd dose: 1 month after 1st dose<br />

3rd dose: 6 months after 1st dose<br />

1st dose: day 0 (day of<br />

vaccination)<br />

2nd dose: 1 month after 1st dose<br />

3rd dose: 6 months after 1st dose<br />

Monovalent hepatitis B vaccines – 2-dose schedule ONLY for adolescents aged 11–15 years<br />

Engerix-B (adult<br />

formulation)<br />

H-B-Vax II (adult<br />

formulation)<br />

11–15<br />

years<br />

11–15<br />

years<br />

Combination hepatitis A/hepatitis B vaccines<br />

Twinrix (720/20) §<br />

Twinrix Junior<br />

(360/10)<br />

1–


months following commencement of the schedule. However, multiple studies<br />

have consistently shown that antibody levels are substantially lower at month 7,<br />

after 3 accelerated doses, than after the standard 3-dose schedule (0, 1, 6 months). 39<br />

Also, some studies, in particular among persons who inject drugs and/or inmates<br />

of correctional facilities, have shown a lower proportion of subjects attaining the<br />

seroprotective antibody level after 3 doses of an accelerated schedule than after<br />

the standard 3-dose schedule. 39,44,45 After the 4th dose of an accelerated schedule,<br />

administered at 12 months, anti-HBs antibody levels are higher or comparable<br />

to those after a standard 3-dose schedule. Hence, it is important for long-term<br />

protection that a 4th dose be administered at 12 months to complete an<br />

accelerated schedule.<br />

Accelerated schedules should only be used for those persons with an imminent<br />

risk of exposure, such as those intending to travel to hepatitis B endemic areas<br />

with a very limited time before departure. As higher seroprotective rates after the<br />

3rd dose of an accelerated 4-dose schedule are seen after the 0, 1, 2, 12 months<br />

schedule than after the 0, 7, 21 days, 12 months schedule, it is recommended that<br />

the latter schedule only be used in exceptional circumstances.<br />

Table 4.5.2: Accelerated hepatitis B vaccination schedules (for persons with<br />

imminent risk of exposure)<br />

Vaccine<br />

Engerix-B<br />

(paediatric<br />

formulation)<br />

Engerix-B<br />

(adult<br />

formulation)<br />

Twinrix<br />

(720/20)<br />

Age of<br />

vaccine<br />

recipient<br />

(years)<br />

Dose<br />

(HBsAg<br />

protein)<br />

Volume<br />

(mL)<br />

Number<br />

of doses<br />


Combination hepatitis A/hepatitis B vaccine schedules<br />

<strong>The</strong> schedules for combination hepatitis A/hepatitis B vaccines are shown in<br />

Table 4.5.1 (and 4.4 Hepatitis A). Three-dose schedules for adults and children<br />

aged


mixed regimens that used two monovalent hepatitis B vaccines from different<br />

manufacturers. 47 As there is only one brand of combination hepatitis A/<br />

hepatitis B vaccine, interchangeability is not relevant. (See also ‘Combination<br />

hepatitis A/hepatitis B vaccine schedules’ in 4.5.4 Vaccines above.)<br />

4.5.7 Recommendations<br />

Infants and young children<br />

<strong>The</strong> recommended hepatitis B vaccine schedule for infants from birth is shown in<br />

Table 4.5.1. A birth dose of monovalent paediatric formulation hepatitis B vaccine<br />

is recommended for all newborn infants. Following this birth dose, 3 doses of a<br />

hepatitis-B-containing combination vaccine (usually provided as DTPa-hepB-<br />

IPV-Hib) are recommended for all children, at 2, 4 and 6 months of age. Thus, a<br />

total of 4 doses of hepatitis B vaccine are provided in the 1st year of life. <strong>The</strong> 1st<br />

dose of a hepatitis B-containing combination vaccine can be given as early as<br />

6 weeks of age. If the 1st dose is given at 6 weeks of age, the next scheduled<br />

doses should still be given at 4 months and 6 months of age.<br />

If an infant has not received a birth dose within the 1st 7 days of life, a primary<br />

3-dose course of a hepatitis B-containing combination vaccine should be given, at<br />

2, 4 and 6 months of age; catch-up of the birth dose is not necessary. Irrespective<br />

of whether a birth dose was given, the infant should not be given the final dose<br />

before 24 weeks of age.<br />

<strong>The</strong> rationale for recommending the birth dose for all newborn infants is not only<br />

to prevent vertical transmission from a mother with chronic hepatitis B infection<br />

(recognising that there may be errors or delays in maternal testing, reporting,<br />

communication or appropriate response), but also to prevent horizontal<br />

transmission to the infant in the first months of life from persons with chronic<br />

hepatitis B infection who are household or other close contacts. 48 <strong>The</strong> birth dose<br />

should be given as soon as the baby is medically stable, and preferably within<br />

24 hours of birth. Every effort should be made to administer the vaccine before<br />

discharge from the obstetric hospital or delivery unit. All newborns of mothers<br />

known to have chronic hepatitis B infection must be given a birth dose of<br />

hepatitis B vaccine and hepatitis B immunoglobulin (HBIG) (see ‘Management of<br />

infants born to mothers who are HBsAg-positive’ below).<br />

Although it is not routinely recommended in Australia, infants or toddlers<br />

who have received a 3-dose schedule of monovalent vaccine (often given<br />

overseas) with doses at birth, 1–2 months of age and ≥6 months of age can also<br />

be considered fully vaccinated. <strong>The</strong> important consideration is that there should<br />

have been an interval of at least 2 months between the 2nd and 3rd doses, and<br />

that the final dose should not be administered before 24 weeks of age (see also<br />

4.5.4 Vaccines above).<br />

218 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Management of infants born to mothers who are HBsAg-positive<br />

Routine antenatal screening of pregnant women for HBsAg is recommended<br />

to enable appropriate management to prevent newborn infants developing<br />

HBV infection (see 4.5.2 Clinical features and 4.5.3 Epidemiology above). 49-51 It also<br />

enables appropriate follow-up and management of mothers who have chronic<br />

HBV infection, identification of the HBV immune status of other household<br />

members, and protection of those who are susceptible to HBV infection.<br />

Infants born to HBsAg-positive mothers should be given HBIG and a dose of<br />

monovalent hepatitis B vaccine on the day of birth, concurrently but in separate<br />

thighs. <strong>The</strong> dose of HBIG is 100 IU, to be given by IM injection. It is preferable to<br />

administer HBIG immediately after birth (preferably within 12 hours of birth and<br />

certainly within 48 hours) as its efficacy decreases markedly if given more than<br />

48 hours after birth.<br />

<strong>The</strong> dose of monovalent hepatitis B vaccine should be given to the infant<br />

preferably within 24 hours of birth, and definitely within 7 days. This regimen<br />

results in seroconversion rates of more than 90% in neonates, even with<br />

concurrent administration of HBIG. Vaccination should not be delayed beyond<br />

7 days after birth, as vaccination alone has been shown to be reasonably effective<br />

in preventing infection, provided it is given early. 52 Three subsequent doses of<br />

a hepatitis B-containing vaccine should be given, at 2, 4 and 6 months of age, so<br />

that the infant receives a total of 4 doses of hepatitis B-containing vaccines.<br />

Anti-HBs antibody and HBsAg levels should be measured in infants born to<br />

mothers with chronic hepatitis B infection 3 to 12 months after completing<br />

the primary vaccine course. Testing should not be performed before 9 months<br />

of age to avoid detection of anti-HBs antibodies from HBIG given at birth. If<br />

anti-HBs antibody levels are adequate (≥10 mIU/mL) and HBsAg is negative,<br />

then the infant is considered to be protected30 (see ‘Serological testing following<br />

hepatitis B vaccination’ below). If the anti-HBs level is


HIV-positive and immunocompromised children<br />

All HIV-positive and immunocompromised children should be age-appropriately<br />

vaccinated against hepatitis B.<br />

HIV-positive children should receive 3 doses of hepatitis B vaccine using an<br />

adult formulation (i.e. double the standard recommended dose for children). In a<br />

limited number of studies, paediatric haemodialysis patients have demonstrated<br />

improved response when given higher doses in a 3-dose schedule. 56,57<br />

For specific hepatitis B recommendations for immunocompromised children, see<br />

3.3.3 Vaccination of immunocompromised persons.<br />

Adolescents<br />

Vaccination of adolescents 10–13 years of age is recommended for all those in<br />

this age group who have not already received a primary course of hepatitis B<br />

vaccine. Refer to your state/territory health authority for further information<br />

about hepatitis B vaccine for this age group (see Appendix 1 Contact details for<br />

<strong>Australian</strong>, state and territory government health authorities and communicable disease<br />

control).<br />

As the risk in <strong>Australian</strong> schools is very low, 58 vaccination of classroom contacts<br />

of hepatitis B cases is seldom indicated. Nevertheless, vaccination of all children<br />

and adolescents should be encouraged.<br />

A 2-dose schedule of hepatitis B vaccine using the adult formulation of either of<br />

the available monovalent vaccines should be considered for adolescents aged<br />

11–15 years who are to receive hepatitis B vaccination (see Table 4.5.1 and 4.5.4<br />

Vaccines above). A 2-dose schedule increases compliance and thus protection in<br />

this age group.<br />

Adolescents who did not receive an age-appropriate completed course of<br />

vaccination should be identified and offered catch-up vaccination, particularly if<br />

they fall into one of the risk categories for hepatitis B infection, discussed under<br />

‘Adults’ below.<br />

Adults<br />

Hepatitis B vaccination is recommended for the following groups of adults<br />

because they are either at a higher risk of acquiring hepatitis B infection and/<br />

or at higher risk of severe disease. Serological testing for previous or chronic<br />

hepatitis B infection may be indicated in many circumstances (see ‘Serological<br />

testing prior to hepatitis B vaccination’ below). See also 3.3 Groups with special<br />

vaccination requirements.<br />

When vaccination against both hepatitis B and hepatitis A is indicated, the<br />

combination hepatitis A/hepatitis B vaccines may be used. See Tables 4.5.1 and<br />

4.5.2 above and ‘Recommendations for the use of combination hepatitis A/<br />

hepatitis B vaccines’ below.<br />

220 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Household or other close (household-like) contacts of persons with hepatitis B<br />

<strong>The</strong>re is a low, but definite, risk of transmission from a person with acute or<br />

chronic hepatitis B to household or other close residential contacts (e.g. students<br />

or asylum seekers sharing residential facilities). This can be reduced by avoiding<br />

contact with blood or other body fluids and not sharing items that may penetrate<br />

the skin (such as combs, nail brushes, toothbrushes and razors). <strong>Immunisation</strong><br />

of susceptible household-like contacts is strongly recommended. This includes<br />

household members of the adoptive family if the adopted child is known to have<br />

chronic hepatitis B infection.<br />

Sexual contacts of persons with hepatitis B<br />

Susceptible sexual partners of persons with acute hepatitis B should be offered<br />

post-exposure HBIG and hepatitis B vaccination; both should be initiated within<br />

14 days of the last sexual contact (see 4.5.11 Public health management of hepatitis B<br />

below and Table 4.5.3). Susceptible partners of those with chronic HBV infection<br />

should also be offered vaccination.<br />

Hepatitis B is relatively common in clients of sexual health services and<br />

vaccination should be offered to susceptible persons at the time of first<br />

attendance.<br />

Susceptible, sexually active men who have sex with men should be vaccinated.<br />

<strong>The</strong> combination hepatitis A/hepatitis B vaccine may be appropriate for men<br />

who have sex with men, if they are not immune to either disease, as they are<br />

at increased risk of both conditions (see ‘Recommendations for the use of<br />

combination hepatitis A/hepatitis B vaccines’ below).<br />

Migrants from hepatitis B endemic countries<br />

Migrants from hepatitis B endemic countries have a higher likelihood of having<br />

been previously infected with hepatitis B and of having a close household contact<br />

with chronic hepatitis B infection. Such persons should be offered testing for<br />

hepatitis B, and vaccination if appropriate. (See also 3.3.8 Vaccination of migrants<br />

to Australia.) Areas of high endemicity, indicated by high seroprevalence of<br />

HBsAg, include most of East and Southeast Asia (except Japan), Pacific island<br />

groups, parts of central Asia and the Middle East, the Amazon Basin, and sub-<br />

Saharan Africa. 59<br />

Aboriginal and Torres Strait Islander people<br />

<strong>The</strong>re is an increased risk of acquiring new HBV infection among Aboriginal<br />

and Torres Strait Islander people compared with other <strong>Australian</strong>s. 17,18 Although<br />

many younger Aboriginal and Torres Strait Islander people, especially children<br />

and adolescents, would have been eligible for, and have received, hepatitis B<br />

vaccination through population-wide vaccination programs, it is recommended<br />

that Aboriginal and Torres Strait Islander people have their risks and vaccination<br />

status for hepatitis B reviewed, be offered testing for previous hepatitis B<br />

infection, and be offered vaccination if non-immune. (See also 3.1 Vaccination for<br />

Aboriginal and Torres Strait Islander people.)<br />

PART 4 VACCINE-PREVENTABLE DISEASES 221<br />

4.5 HEPATITIS B


Adult haemodialysis patients and patients with severely impaired renal function in whom<br />

dialysis is anticipated<br />

Dialysis patients, and patients with severely impaired renal function in whom<br />

dialysis is anticipated, may be at increased risk of acquiring hepatitis B infection<br />

and also respond less well to vaccination. <strong>The</strong>se patients should be given a larger<br />

than usual dose of hepatitis B vaccine.<br />

Adult haemodialysis or pre-dialysis patients should be given either:<br />

• 1.0 mL of Engerix-B adult formulation (20 µg) in each arm at each schedule<br />

point (i.e. effectively giving a double dose on each occasion) in a 4-dose<br />

schedule at 0, 1, 2 and 6 months; 60 or<br />

• a single dose of H-B-Vax II dialysis formulation (40 µg) on each occasion in a<br />

3-dose schedule at 0, 1 and 6 months.<br />

Solid organ and haematopoietic stem cell transplant recipients<br />

If seronegative for hepatitis B, solid organ transplant recipients should be<br />

vaccinated before transplantation as they may be at increased risk of infection<br />

from the transplanted organ. 61 Haematopoietic stem cell transplant recipients<br />

should be revaccinated following transplantation, due to the loss of immune<br />

memory that often follows the transplant procedure. (See also 3.3.3 Vaccination of<br />

immunocompromised persons.)<br />

HIV-positive adults and other immunocompromised adults<br />

HIV-positive adults, and other immunocompromised adults, may be at increased<br />

risk of acquiring hepatitis B infection and also respond less well to vaccination.<br />

Limited studies in HIV1-positive adults have demonstrated an improved and<br />

accelerated serological response to a schedule that consists of 4 double doses,<br />

comprising two injections of the standard adult dose (using Engerix-B) on each<br />

occasion, at times 0, 1, 2 and 6 months. 62,63<br />

Persons with chronic liver disease and/or hepatitis C<br />

Hepatitis B vaccination is recommended for those in this category who are<br />

seronegative for hepatitis B, because of the risk of severe liver disease following<br />

infection with hepatitis B. 64<br />

Persons who inject drugs<br />

Persons who inject drugs should be tested, and be vaccinated if they have not<br />

previously been infected with HBV.<br />

Recipients of certain blood products<br />

Screening of all blood donors for HBV using HBsAg and nucleic acid<br />

amplification tests has greatly decreased the incidence of transfusion-related<br />

hepatitis B virus infection. Since 2010, nucleic acid testing has been introduced<br />

nationally to improve detection of hepatitis B infection in donated blood, mainly<br />

through reduction of the infectious window period when acute hepatitis B<br />

infection may not be detected using HBsAg, but also through detecting persons<br />

222 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


with occult hepatitis B infection. This further reduces the residual risk of<br />

hepatitis B transmission through transfusion in Australia, from approximately 1<br />

in 739 000 to less than 1 in 1 million. 65 However, persons with clotting disorders<br />

who receive blood product concentrates, persons with recurrent transfusion<br />

requirements, and persons with underlying immunocompromise66 have an<br />

elevated risk of hepatitis B virus infection, and should therefore be vaccinated.<br />

Persons with developmental disabilities<br />

Vaccination is recommended for persons who attend either residential or nonresidential<br />

day-care facilities for persons with developmental disabilities. This<br />

is due to the high prevalence of markers indicating past or current infection in<br />

persons in these settings, including an HBsAg prevalence of >10%. 67-69<br />

Inmates of correctional facilities<br />

Inmates are at increased risk of hepatitis B infection because of the prevalence<br />

of chronic hepatitis B among inmates, and the potential for unprotected sexual<br />

intercourse, injecting drug use and amateur tattooing in correctional facilities.<br />

<strong>The</strong>refore, they should be offered the opportunity to be screened for hepatitis B<br />

upon incarceration, as part of the preventive health program for blood-borne<br />

viruses, and vaccinated if susceptible.<br />

Sex industry workers<br />

Sex industry workers are one of the population groups at higher risk of HBV<br />

infection. <strong>The</strong>y have been specifically identified as an important population on<br />

which to focus for the prevention of hepatitis B transmission. 70 <strong>The</strong>y are at a<br />

particularly high risk if they engage in unprotected sex.<br />

Persons at occupational risk<br />

<strong>The</strong> risk to persons in certain occupations differs considerably from setting to<br />

setting in different parts of Australia. However, it is recommended that all staff<br />

directly involved in patient care and/or the handling of human blood or tissue<br />

should be vaccinated. In addition, standard precautions against exposure to<br />

blood or body fluids should be used as a matter of routine. 71<br />

Other occupations where the risk of acquiring hepatitis B is increased include:<br />

• police, members of the armed forces, emergency services staff and staff of<br />

correctional facilities; these persons should be vaccinated if they are assigned<br />

to duties that may involve exposure to blood/body fluids<br />

• funeral workers, embalmers and other workers who have regular contact<br />

with human tissue, blood or body fluids and/or used needles or syringes<br />

• staff involved in both residential and non-residential care of persons with<br />

developmental disabilities, due to the high prevalence of markers of past or<br />

current infection in persons in this setting67-69 • workers who perform skin penetration procedures (e.g. tattooists, bodypiercers).<br />

PART 4 VACCINE-PREVENTABLE DISEASES 223<br />

4.5 HEPATITIS B


Staff of child day-care centres will normally be at minimal risk of hepatitis B. If<br />

advice on risk is sought, the enquiry should be directed to the local public health<br />

authority.<br />

Contact sports generally carry a low risk of hepatitis B infection. However, ageappropriate<br />

hepatitis B vaccination is recommended.<br />

Travellers to hepatitis B endemic areas<br />

Persons travelling to regions of intermediate or high endemicity, either long-term<br />

or for frequent short terms, or who are likely to undertake activities that increase<br />

their risks of exposure to HBV during travel, should be vaccinated. 59 (See also 3.2<br />

Vaccination for international travel.)<br />

Recommendations for the use of combination hepatitis A/hepatitis B vaccines<br />

Combination hepatitis A/hepatitis B vaccines should be considered for<br />

susceptible persons in whom both hepatitis A and hepatitis B vaccines are<br />

recommended, including:<br />

• travellers to, and expatriates living in, moderately to highly endemic areas<br />

for hepatitis A and B<br />

• persons whose lifestyle puts them at increased risk of hepatitis A and<br />

hepatitis B (sexually active men who have sex with men, sex industry<br />

workers, persons who inject drugs and inmates of correctional facilities)<br />

• persons who attend or work at residential or non-residential facilities for<br />

people with developmental disabilities<br />

• persons with occupational risks of exposure to both hepatitis A and hepatitis B<br />

• persons with chronic liver disease and/or hepatitis C<br />

• solid organ transplant liver recipients or solid organ transplant recipients<br />

who have chronic liver disease (see Table 3.3.2 Recommendations for<br />

vaccinations for solid organ transplant (SOT) recipients).<br />

If a combination hepatitis A/hepatitis B vaccine is not available, monovalent<br />

hepatitis A and hepatitis B vaccines can be administered simultaneously (in<br />

separate syringes at separate sites) (see ‘Interchangeability of hepatitis B<br />

vaccines’ above).<br />

See 4.5.7 Recommendations above and 4.4 Hepatitis A for more details. See also 3.3<br />

Groups with special vaccination requirements.<br />

Booster doses<br />

Booster doses of hepatitis B vaccine (after completion of a primary course by a<br />

recommended schedule) are not recommended for immunocompetent persons.<br />

This applies to children and adults, including healthcare workers and dentists. 72-78<br />

This is because there is good evidence that a completed primary course of<br />

hepatitis B vaccination provides long-lasting protection. Even though vaccineinduced<br />

antibody levels may decline with time and may become undetectable,<br />

224 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


immune memory persists and is thought to result in a protective immune<br />

response on re-exposure. 79 However, booster doses are recommended for persons<br />

who are immunocompromised, in particular those with either HIV infection or<br />

renal failure. <strong>The</strong> time for boosting in such persons should be decided by regular<br />

monitoring of anti-HBs levels at 6- to 12-monthly intervals. 72<br />

Serological testing prior to hepatitis B vaccination<br />

Routine antenatal screening of all pregnant women for HBsAg is recommended<br />

to allow appropriate measures to be implemented to prevent newborn infants<br />

developing chronic HBV infection49-51 (see ‘Management of infants born to<br />

mothers who are HBsAg-positive’ above).<br />

Serological testing for evidence of past (or current) hepatitis B infection prior<br />

to vaccination may be warranted for certain older children, adolescents<br />

and adults. This is particularly so for those at increased risk of acquiring<br />

hepatitis B infection, such as persons who inject drugs, sex industry workers,<br />

immunocompromised persons, and those living in communities with higher<br />

prevalence of HBV, including migrant communities and Aboriginal and Torres<br />

Strait Islander people. Serological testing enables identification of persons who<br />

were infected by HBV, to facilitate timely appropriate clinical management and<br />

prevention of onward transmission, hence reducing population impact of HBV<br />

infection. Testing also identifies those who are susceptible to HBV infection and,<br />

as such, should be offered vaccination if they continue to have a high exposure<br />

risk (see 4.5.7 Recommendations above). 70 Testing for immunity to hepatitis A<br />

infection (and vaccination of susceptible at-risk persons with combination<br />

hepatitis A/hepatitis B vaccines) may also be indicated for some population<br />

groups at increased risk of hepatitis A exposure (see 4.4 Hepatitis A).<br />

Interpretation of the results of serological testing may be enhanced by discussion<br />

with the laboratory that performed the test, ensuring that relevant clinical<br />

information is provided.<br />

Serological testing following hepatitis B vaccination<br />

Anti-HBs antibody and HBsAg levels should be measured in infants born to<br />

mothers with chronic hepatitis B infection 3 to 12 months after completing the<br />

primary vaccine course (for more information see ‘Management of infants born<br />

to mothers who are HBsAg-positive’ above).<br />

Other than for infants born to mothers with chronic hepatitis B infection, postvaccination<br />

serological testing is recommended 4 to 8 weeks after completion of<br />

the primary course for persons in the following categories:<br />

• those at significant occupational risk (e.g. healthcare workers whose work<br />

involves frequent exposure to blood and body fluids)<br />

• those at risk of severe or complicated HBV disease (e.g. persons who are<br />

immunocompromised, and persons with pre-existing liver disease not related<br />

to hepatitis B)<br />

PART 4 VACCINE-PREVENTABLE DISEASES 225<br />

4.5 HEPATITIS B


• those in whom a poor response to hepatitis B vaccination may occur (e.g.<br />

haemodialysis patients, persons with bleeding disorders vaccinated via the<br />

SC route)<br />

• sexual partners and household, or other close household-like, contacts of<br />

persons who are infected with hepatitis B. 30<br />

For these individuals, if adequate anti-HBs levels (≥10 mIU/mL) are not reached<br />

on serological testing 4 to 8 weeks after the 3rd dose, the possibility of HBV<br />

infection, including chronic HBV infection, should be investigated by testing for<br />

serological markers, including HBsAg and anti-HBc antibodies. In select cases<br />

in which hepatitis B infection is suspected, HBV nucleic acid testing may also be<br />

indicated, and expert advice regarding further management should be sought.<br />

If there are no markers of HBV infection, the individual should be managed<br />

as a non-responder to hepatitis B vaccination (see ‘Non-responders to primary<br />

vaccination’ below).<br />

If persons who are at significant risk of hepatitis B (such as healthcare workers)<br />

were not tested for anti-HBs within 4 to 8 weeks after completion of the<br />

documented primary course, they should still undergo serological testing to<br />

ensure immunity. If, on testing, they have an anti-HBs level of


to suggest that a higher proportion of subjects would respond with these higherdose<br />

regimens. 80-82<br />

For HBsAg-negative healthcare workers who are non-responders to a primary<br />

course of vaccination and to subsequent additional IM doses (≥5 doses in total),<br />

some small observational studies report that some individuals may respond<br />

to the vaccine administered intradermally. 83-85 Engerix-B (0.25 mL [5 μg] per<br />

dose) was used in these studies, giving up to 4 doses. 83 Younger age and longer<br />

duration (≥6 months) since the last IM dose may be associated with greater<br />

probability of response. 84 If an intradermal dose(s) is given, it is recommended<br />

that the anti-HBs levels be measured before each subsequent dose to assess for<br />

seroconversion.<br />

Persistent non-responders should be informed that they should be considered<br />

not protected against hepatitis B and should minimise exposures. <strong>The</strong>y should<br />

also be informed about the need for HBIG within 72 hours of parenteral or<br />

mucosal exposure to HBV (see Table 4.5.3).<br />

4.5.8 Pregnancy and breastfeeding<br />

Hepatitis B vaccine is not routinely recommended for pregnant or breastfeeding<br />

women. However, the WHO states that neither pregnancy nor breastfeeding is a<br />

contraindication to the use of this vaccine. 86<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.5.9 Contraindications<br />

<strong>The</strong> only absolute contraindications to hepatitis B vaccines are:<br />

• anaphylaxis following a previous dose of any hepatitis B vaccine<br />

• anaphylaxis following any vaccine component.<br />

In particular, hepatitis B vaccines are contraindicated in persons with a history of<br />

anaphylaxis to yeast.<br />

4.5.10 Adverse events<br />

Extensive experience indicates that the birth dose of hepatitis B vaccine is<br />

very well tolerated by newborn infants. It does not interfere with either the<br />

establishment or maintenance of breastfeeding, and it is not associated with<br />

an increased risk of either fever, medical investigation for sepsis, or serious<br />

outcomes in newborns who were vaccinated compared with the unvaccinated. 87-89<br />

Adverse events after hepatitis B vaccination are transient and minor, and include<br />

soreness at the injection site (5%), fever (usually low grade; 2–3%), nausea,<br />

dizziness, malaise, myalgia and arthralgia. Fever can be expected in some<br />

neonates following immunisation with hepatitis B vaccine (0.6–3.7%).<br />

Anaphylaxis has been reported very rarely in adults, notably in yeast-sensitive<br />

individuals. 90 Although various adverse events such as demyelinating diseases,<br />

PART 4 VACCINE-PREVENTABLE DISEASES 227<br />

4.5 HEPATITIS B


Guillain-Barré syndrome and arthritis have been reported, there is no evidence of<br />

a causal relationship with hepatitis B vaccination. 90,91<br />

<strong>The</strong> World Health Organization Global Advisory Committee on Vaccine Safety<br />

states that ‘multiple studies and review panels have concluded that there is no<br />

link between MS [multiple sclerosis] and hepatitis B vaccination’. 92,93<br />

<strong>The</strong> vaccine produces neither therapeutic effects nor adverse events in persons<br />

with chronic HBV infection. It is also safe, though of no additional benefit, in<br />

those already immune to hepatitis B through past natural infection.<br />

4.5.11 Public health management of hepatitis B<br />

Acute hepatitis B and newly identified chronic hepatitis B are notifiable diseases<br />

in all states and territories in Australia.<br />

Further instructions about the public health management of hepatitis B,<br />

including management of cases of acute hepatitis B and newly identified chronic<br />

hepatitis B, and their contacts, should be obtained from state/territory public<br />

health authorities (see Appendix 1 Contact details for <strong>Australian</strong>, state and territory<br />

government health authorities and communicable disease control).<br />

Following significant exposure (percutaneous, ocular or mucous membrane) to<br />

blood or to potentially blood-contaminated secretions, where feasible, the source<br />

individual should be tested for HBsAg as soon as possible.<br />

If the person exposed has not been previously vaccinated against hepatitis B,<br />

their anti-HBs level, and anti-HBc and HBsAg status, should be determined<br />

immediately. If the person exposed is anti-HBs and anti-HBc negative (nonimmune)<br />

and the source is either HBsAg-positive or cannot be identified and<br />

tested rapidly, a single dose of HBIG should be administered according to<br />

the recommendations in Table 4.5.3. <strong>The</strong> dose of HBIG is 100 IU for children<br />

weighing up to 30 kg (about 5 years of age) and 400 IU for all others. Hepatitis B<br />

vaccine must also be given as soon as possible, with further doses as<br />

recommended in Table 4.5.3.<br />

For previously vaccinated persons exposed to either an HBsAg-positive source<br />

or a source whose hepatitis B status cannot be determined, post-exposure<br />

prophylaxis is not necessary if there was a documented protective response (anti-<br />

HBs level ≥10 mIU/mL) at any time after vaccination. If the response to previous<br />

vaccination is unknown, the anti-HBs level should be determined as quickly as<br />

possible. If the anti-HBs level is


Table 4.5.3: Post-exposure prophylaxis for non-immune persons exposed to a<br />

HBsAg-positive source<br />

Type of exposure Hepatitis B immunoglobulin Vaccine<br />

Perinatal<br />

(exposure of<br />

babies during and<br />

after birth)*<br />

Percutaneous,<br />

ocular or mucous<br />

membrane<br />

100 IU, by<br />

IM injection<br />

400 IU, by<br />

IM injection<br />

100 IU,<br />

if body<br />

weight<br />


4.5.12 Variations from product information<br />

<strong>The</strong> product information for Infanrix hexa states that this vaccine is indicated<br />

for primary immunisation of infants from the age of 6 weeks. <strong>The</strong> ATAGI<br />

recommends that this vaccine may also be used for catch-up of the primary<br />

schedule in children


4.6 HUMAN PAPILLOMAVIRUS<br />

4.6.1 Virology<br />

Human papillomaviruses (HPVs) are small, non-enveloped viruses with circular<br />

double-stranded DNA. HPVs infect and replicate primarily within cutaneous<br />

and mucosal epithelial tissues.<br />

More than 100 HPV genotypes have been identified based on sequence variations<br />

in the major genes. <strong>The</strong>y differ in their preferred site of infection; approximately<br />

40 HPV types infect the anogenital tract. Some HPV types, including types 16,<br />

18, 31, 33, 35, 45, 52 and 58, are designated as ‘high-risk’, as they are causally<br />

associated with the development of cancer. Other HPV types, including types<br />

6, 11, 40, 42, 43, 44, 54, 61, 70, 72, 81 and 89, have been classified as ‘low-risk’<br />

and are predominantly associated with non-malignant lesions, such as genital<br />

warts. <strong>The</strong> other types are uncommon and their associations with disease are<br />

undetermined, but they are not currently believed to be significant causes of<br />

cancer. 1,2<br />

4.6.2 Clinical features<br />

Transmission of anogenital HPV occurs primarily through sexual intercourse;<br />

however, virus transmission can less commonly occur following non-penetrative<br />

sexual contact. 3 Perinatal transmission of HPV can cause laryngeal infection in<br />

infants, rarely resulting in recurrent respiratory papillomatosis. 4 HPV infection is<br />

often subclinical, but, dependent upon the infecting HPV genotype, may result<br />

in lesions that include cutaneous warts, genital warts, respiratory papillomatosis<br />

(low-risk HPV types), and dysplasias and cancers of the cervix, vulva, vagina,<br />

penis, anus, and the oral cavity and oropharynx (high-risk HPV types). Most<br />

genital HPV infections are cleared (no longer detectable via HPV DNA testing)<br />

within 12 to 24 months. In about 3 to 10% of infections, the virus persists. Persons<br />

with persistent HPV infection constitute the at-risk group for development of<br />

HPV-associated cancers. 5-7<br />

<strong>The</strong> causal link between persistent cervical HPV infection and cervical cancer is<br />

well established. <strong>The</strong> strength of association between HPV infection and other<br />

cancers varies by site and oncogenic HPV type. 8<br />

Cellular changes that occur in the cervix as a result of HPV infection are referred<br />

to as cervical intraepithelial neoplasia (CIN). <strong>The</strong> majority of these changes<br />

regress, but a minority will progress to cervical cancer. Malignant transformation<br />

in the cervix usually occurs 10 to 20 years following infection with high-risk HPV<br />

types, but has been reported to occur in less than 2 years. 9<br />

<strong>The</strong> clinical features of other HPV-associated cancers and their precursor lesions<br />

in the anogenital region and oropharynx vary, and also depend on the anatomical<br />

site. <strong>The</strong> process of progression of HPV-associated precursor lesions to cancers<br />

in these sites is less well understood than the process in the cervix. Anogenital<br />

warts may present as painless lumps, or with local tenderness, itching or<br />

PART 4 VACCINE-PREVENTABLE DISEASES 231<br />

4.6 HUMAN PAPILLOMAVIRUS


leeding. Recurrent respiratory papillomatosis is a potentially fatal condition that<br />

usually occurs in childhood, characterised by multiple warty excrescences on the<br />

mucosal surface of the respiratory tract. 10<br />

4.6.3 Epidemiology<br />

Infection with HPV is very common in both men and women, with initial<br />

infection occurring close to the time of sexual debut. It is estimated that up to<br />

79% of the general population will be infected with at least one genital type<br />

of HPV at some time in their lives. 11,12 A greater number of sexual partners is<br />

consistently found to be associated with an increased risk of HPV acquisition. 12<br />

HPV infection rates differ between geographic regions, and estimated<br />

population prevalence of HPV also varies depending on the anatomical site and<br />

the lesions sampled. About two-thirds of <strong>Australian</strong> women aged 15–20 years<br />

participating in cervical screening had HPV DNA detected in cervical samples<br />

collected for cytology. 13<br />

Certain population subgroups are identified to be at increased risk of<br />

HPV infection and HPV-associated diseases, compared with the general<br />

population. Infection with multiple HPV genotypes and longer time to clear<br />

infection are commonly observed in men who have sex with men (MSM). 14-16<br />

In addition, the prevalence of high-risk HPV types is significantly higher in<br />

HIV-positive MSM than in MSM who are HIV-negative. 14 Persons who are<br />

immunocompromised (due to disease or medical treatment) are at increased<br />

risk of HPV-related disease. 12<br />

In a serosurvey conducted in Australia in 2006, 24% of females and 18% of males<br />

aged 0–69 years were seropositive to at least one of the four HPV types 6, 11, 16<br />

and 1817 – noting that fewer than 60% of women, and an even lower proportion<br />

of men, who are infected with HPV develop antibodies. 18-20 <strong>The</strong> onset age of<br />

seropositivity for HPV in this study was 10–14 years in females and 15–19 years<br />

in males. <strong>The</strong> average age of sexual debut for both males and females in Australia<br />

was 16 years, as reported in 2000–2002. 21 A more recent national survey in 2008<br />

reported that about 80% of senior secondary school children (aged approximately<br />

15–19 years) acknowledged having engaged in some form of sexual activity that<br />

may transmit HPV. 22<br />

Persistent HPV infection is the necessary precursor for the development<br />

of all cervical cancers. 23 Worldwide, approximately 70% of cervical cancers<br />

contain HPV-16 DNA and 16% contain HPV-18 DNA. 24,25 <strong>Australian</strong> data<br />

indicate that HPV-16 and HPV-18 are responsible for approximately 60% and<br />

20%, respectively, of cervical cancers, and 37% and 8%, respectively, of highgrade<br />

cervical abnormalities. 26,27 In Australia, cervical cancer ranked 22nd in<br />

the overall cancer disease burden in 2008 and now occurs predominantly in<br />

women unscreened or under-screened through the National Cervical Screening<br />

Program. 28,29 In 2007, the age-standardised incidence rate of cervical cancer in<br />

Australia was 6.8 per 100 000, and the mortality rate was 1.8 deaths per 100 000<br />

232 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


women. <strong>The</strong> prevalence of high-risk HPV types 16 and 18, detected when<br />

cervical samples collected for cytology were tested for HPV DNA, was similar<br />

between Indigenous and non-Indigenous women. 13 However, the incidence rate<br />

of cervical cancer in Aboriginal and Torres Strait Islander women is almost 3<br />

times higher than in non-Indigenous <strong>Australian</strong> women, an indication of lower<br />

participation rates in cervical screening programs by Indigenous <strong>Australian</strong>s and<br />

greater prevalence of cofactors for cervical cancer such as smoking, earlier and<br />

more pregnancies, and lower socioeconomic status. 13,28,30 Indigenous women are<br />

5 times more likely to die from cervical cancer than non-Indigenous women. 28<br />

Also, <strong>Australian</strong>s in remote and very remote areas have 1.5 times higher cervical<br />

cancer incidence than those living in major cities. 28<br />

<strong>The</strong> proportion of cancers of other anogenital sites that is attributable to HPV<br />

ranges from approximately 40% for vulval cancers to approximately 85% for<br />

anal cancers. More than 85% of these HPV-associated cancers have evidence of<br />

infection due to the high-risk HPV types 16 and 18. 31-36<br />

In Australia in 2007, incidence rates of vulval and vaginal cancers in women were<br />

2.6 per 100 000 (n=276) and 0.65 per 100 000 (n=69), respectively. 28 <strong>The</strong> incidence<br />

rate of penile cancer was 0.8 per 100 000. <strong>The</strong> age-standardised incidence rate<br />

for anal cancer was 1.3 per 100 000; however, a slightly higher incidence was<br />

observed in females than in males. Overall, anal cancer incidence has been<br />

steadily increasing over the past few decades; however, the increase has been<br />

greater in males than in females. 31,34 <strong>The</strong> mortality rates for vulval, vaginal, penile<br />

and anal cancers were all less than 0.6 per 100 000. 28<br />

MSM have a significantly higher incidence of high-grade anal intraepithelial<br />

neoplasia and anal cancer than the general population. Overseas studies have<br />

found a greater than 30-fold higher incidence of anal cancer in MSM than in<br />

other men. 37,38<br />

<strong>The</strong>re is wide variability in the reported proportions of oropharyngeal cancers<br />

associated with HPV, ranging from 12% to 63%, and a lower proportion of oral<br />

cancers. 39-41 Of the cancers at these sites that are HPV-positive, HPV-16 and/or<br />

HPV-18 account for more than 85%. In Australia, similar to the United States and<br />

other western countries, there has been a steady increase in the burden of HPVpositive<br />

oropharyngeal cancers (mainly attributable to cancers of the base of the<br />

tongue and tonsils) over the past few decades. 31,39,42-45<br />

<strong>The</strong> population incidence of benign HPV-associated lesions, such as anogenital<br />

warts, is much higher than the incidence of HPV-associated cancers. In Australia,<br />

the estimated annual incidence of anogenital warts in 2000–2006 was 206 per<br />

100 000 in males and 231 per 100 000 in females. <strong>The</strong> age group of peak incidence<br />

was 25–29 years for men (rate 740 per 100 000) and 20–24 years for women (rate<br />

861 per 100 000). 46 In Australia, 4.0% of men and 4.4% of women aged 16–59<br />

years reported ever being diagnosed with genital warts, 47 and the estimated<br />

cumulative lifetime risk of genital warts was 10%. 48,49 <strong>The</strong> estimated incidence of<br />

PART 4 VACCINE-PREVENTABLE DISEASES 233<br />

4.6 HUMAN PAPILLOMAVIRUS


anogenital warts in MSM is about 10 times higher than in the general population,<br />

with a third of HIV-negative MSM reporting a history of these lesions. 46,50 HPV<br />

types 6 and 11 are associated with 90% of genital warts. 51,52<br />

Recurrent respiratory papillomatosis is a rare (incidence approximately 3.5 per<br />

100 000) and predominantly childhood disease that is associated with HPV types<br />

6 and 11 in 100% of cases. 52-54<br />

In 2007, the HPV Vaccination Program, funded under the NIP, was introduced.<br />

This initially included universal vaccination of girls aged approximately<br />

12–13 years, delivered through an ongoing school-based program. It also<br />

included a catch-up program for females up to 26 years of age, which ceased at<br />

the end of 2009. In <strong>2013</strong>, the program will be extended to include HPV vaccine<br />

for boys aged approximately 12–13 years, together with a 2-year catch-up<br />

program for Year 9 boys. Although the impact of HPV vaccination on cancer<br />

incidence will take decades to occur, early surveillance data have shown an<br />

impact on the incidence of genital warts and CIN in the years following the<br />

introduction of the female vaccination program. 55-58 A study including eight<br />

sexual health centres showed a 59% decrease in the proportion of vaccineeligible<br />

female first-time clinic attendees diagnosed with genital warts. 56 This<br />

study also demonstrated that vaccination of females results in some herd<br />

immunity benefits to males, with a significant decline in the diagnosis of genital<br />

warts observed in unvaccinated males of the same age. 55,56,58 In addition to<br />

reduction in genital warts, Victorian data have demonstrated a 48% decline in<br />

the incidence of high-grade cervical abnormalities in girls aged


• Gardasil – CSL Limited/Merck & Co Inc (recombinant protein<br />

particulate [VLP] vaccine containing the major capsid [L1] protein<br />

of HPV types 6, 11, 16 and 18; 4vHPV). Each 0.5 mL monodose<br />

vial or pre-filled syringe contains 20 µg HPV-6 L1 protein, 40 µg<br />

HPV-11 L1 protein, 40 µg HPV-16 L1 protein and 20 µg HPV-18<br />

L1 protein, adsorbed onto 0.225 mg of aluminium as aluminium<br />

hydroxyphosphate sulphate; 0.780 mg L-histidine; 50 µg polysorbate<br />

80; 35 µg sodium borate. May also contain yeast proteins.<br />

<strong>The</strong> 2vHPV and 4vHPV vaccines have been assessed in females in a number<br />

of international clinical trials. When given as a 3-dose series, HPV vaccines<br />

elicit a neutralising antibody level many times higher than the level observed<br />

following natural infection. 61,62 Overall, seroconversion occurs in 97 to 100%<br />

of those vaccinated. 63-65 In women who are naïve to HPV types 16 and 18 prior<br />

to vaccination, both vaccines are highly effective at preventing type-specific<br />

persistent infection and related cervical disease (approximately 90 to 100%). 66-71<br />

<strong>The</strong> 4vHPV vaccine also has established efficacy (100%; 95% CI: 94–100%)<br />

against external anogenital and vaginal lesions (genital warts, and vulval,<br />

vaginal, perineal and perianal dysplasias) associated with HPV types 6, 11,<br />

16 or 18 in women.<br />

In women who are vaccinated irrespective of their baseline HPV status (i.e.<br />

women who may have pre-existing HPV infection), vaccine efficacy is lower than<br />

observed in HPV-naïve women, indicating reduced vaccine effectiveness among<br />

women who are already sexually active. This is because both HPV vaccines are<br />

prophylactic vaccines (i.e. preventing primary HPV infection). Vaccination will<br />

not treat an existing HPV infection or prevent disease that may be caused by an<br />

existing HPV vaccine-type infection. 63,72,73 However, vaccination may still provide<br />

benefit for sexually active women by protecting them against new infections with<br />

other vaccine-preventable HPV types.<br />

<strong>The</strong> efficacy of 4vHPV in males aged 16–26 years has been demonstrated<br />

in one clinical trial. 74 Vaccination was greater than 85% protective against<br />

persistent anogenital infection and external genital lesions due to vaccine HPV<br />

types among HPV-naïve participants. Among HPV-naïve MSM participants<br />

within the clinical trial, vaccine efficacy was 95% against intra-anal HPV<br />

infection and 75% against high-grade anal intraepithelial neoplasia from<br />

vaccine HPV types. Efficacy of 2vHPV vaccine in males has not been assessed<br />

to date; however, the vaccine has demonstrated safety and immunogenicity in<br />

males aged 10–18 years. 75<br />

<strong>The</strong>re is some evidence of HPV vaccine providing some cross-protection to<br />

disease due to other HPV types in women: 4vHPV vaccine against cervical<br />

PART 4 VACCINE-PREVENTABLE DISEASES 235<br />

4.6 HUMAN PAPILLOMAVIRUS


disease due to HPV types 31 and 4576 and 2vHPV vaccine against cervical disease<br />

due to HPV types 31, 33, 45 and 51. 77 However, the level of protection is less than<br />

for the vaccine HPV types and the durability of any such protection is unknown.<br />

Efficacy of HPV vaccines in females or males 9 years of age) were greater than those in adult women and<br />

men, in whom clinical efficacy has been demonstrated for both the 4vHPV and<br />

2vHPV vaccines.<br />

It is not certain how long immunity following HPV vaccination persists, or<br />

whether a booster dose after the primary course will ever be required. However,<br />

long-term population-based follow-up studies to assess this are underway. 78<br />

In clinical trials, vaccine efficacy has been demonstrated up to at least 5 years<br />

for 4vHPV vaccine and 9.4 years for 2vHPV vaccine in women, with no<br />

breakthrough disease due to vaccine HPV types. 61,79,80<br />

Variations in vaccination schedules for both HPV vaccines are being assessed<br />

in clinical trials. A recent study showed a lesser immune response in a schedule<br />

with a dose interval of 12 months between each of the 3 doses of 4vHPV vaccine<br />

compared with schedules with dose intervals of 6 months or less between each of<br />

the doses. 81 However, a recent study of 2vHPV following an alternative schedule<br />

(0, 1 and 12 months) demonstrated that the immunogenicity of vaccine HPV<br />

types was non-inferior following this schedule, compared with the standard<br />

schedule (measured 1 month after the final dose). 82 Two-dose schedules of both<br />

2vHPV and 4vHPV are also being studied. 83,84<br />

4.6.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 85 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

4.6.6 Dosage and administration<br />

<strong>The</strong> dose of both HPV vaccines is 0.5 mL to be given by IM injection.<br />

<strong>The</strong> primary vaccination course for both HPV vaccines consists of 3 doses.<br />

<strong>The</strong> recommended schedule for the 2vHPV vaccine is at times 0 (the day the<br />

1st dose is given), 1 and 6 months. <strong>The</strong> 2vHPV vaccine is registered for use in<br />

females aged 10–45 years. <strong>The</strong> 2vHPV vaccine is not registered for use in males<br />

of any age.<br />

<strong>The</strong> recommended schedule for the 4vHPV vaccine is at times 0 (the day the<br />

1st dose is given), 2 and 6 months. <strong>The</strong> 4vHPV vaccine is registered for use in<br />

females aged 9–45 years and in males aged 9–26 years. However, there are no<br />

theoretical concerns that the efficacy or safety of 4vHPV vaccine in males up<br />

to the age of 45 years will differ significantly from females of the same age or<br />

younger males.<br />

236 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


If scheduled doses have been missed, there is no need to repeat earlier doses.<br />

<strong>The</strong> missed dose(s) should be given as soon as is practicable, making efforts to<br />

complete doses within 12 months.<br />

Where vaccines have been administered at less than the minimum intervals (see<br />

Table 2.1.12 Catch-up schedule for persons ≥10 years of age (for vaccines recommended<br />

on a population level)), contact your state or territory health department for<br />

guidance. See also Chief Medical Officer Guidance available at www.health.gov.<br />

au/internet/immunise/publishing.nsf/Content/cmo-full-advice-hpv-cnt.<br />

Co-administration with other vaccines<br />

Both HPV vaccines can be given concomitantly with reduced antigen content<br />

diphtheria-tetanus-acellular pertussis (dTpa) or diphtheria-tetanus-acellular<br />

pertussis-inactivated poliomyelitis vaccine (dTpa-IPV), and hepatitis B vaccine<br />

(monovalent). 86-90 <strong>The</strong>re are no clinical data regarding concomitant administration<br />

of either HPV vaccine with varicella vaccine, but there are no theoretical concerns<br />

about safety or efficacy of the vaccines if they are given simultaneously, using<br />

different injection sites.<br />

Interchangeability of human papillomavirus vaccines<br />

<strong>The</strong>re are currently no clinical data available on the interchangeability of the<br />

two HPV vaccines. However, from first principles, acceptable antibody levels<br />

and protection against HPV-16 and 18 (the types that are shared by both these<br />

vaccines and that are the dominant causes of cervical cancer) would be expected<br />

following a combination schedule.<br />

It is recommended that an HPV vaccination course commenced with one vaccine<br />

should, wherever possible, be completed with that vaccine and according to its<br />

recommended schedule.<br />

Where the course includes a combination of the two HPV vaccines, either<br />

inadvertently or because of an adverse event following one vaccine, the person<br />

is considered to be fully immunised against HPV-16 and 18 disease if a total of<br />

3 doses of HPV vaccine have been given, provided that the minimum interval<br />

requirements between the doses are satisfied. Every effort should be made to<br />

complete a 3-dose schedule for effective protection against disease due to each of<br />

the vaccine HPV types.<br />

4.6.7 Recommendations<br />

Neither HPV vaccine is registered or recommended for use in children


Children and adolescents aged 9–18 years<br />

HPV vaccine is recommended for females 9–18 years of age. <strong>The</strong> optimal age<br />

for administering the HPV vaccine is approximately 11–13 years, as most<br />

females in this age group would not have commenced sexual activity and so<br />

would be naïve to all HPV types. Vaccination only provides protection against<br />

vaccine-type disease if the vaccine is delivered prior to acquisition of that HPV<br />

type. <strong>The</strong>refore, the decision to vaccinate older adolescent females, who may<br />

have already commenced sexual activities, should follow an assessment of the<br />

potential benefits, based on their likely previous HPV exposure and future risks<br />

of HPV exposure.<br />

Either of the HPV vaccines can be used for adolescent females. <strong>The</strong> 2vHPV<br />

vaccine is only registered for use in girls ≥10 years of age.<br />

Adults aged ≥19 years<br />

Vaccination of all women in this age group is not routinely recommended, as<br />

many are likely to have been exposed to one or more vaccine HPV types through<br />

sexual activity (see 4.6.3 Epidemiology above).<br />

However, some adult females may gain an individual benefit from HPV<br />

vaccination. <strong>The</strong> decision to vaccinate older females should take into account<br />

their likelihood of previous exposure to HPV and their future risks of HPV<br />

exposure.<br />

Males<br />

<strong>The</strong> 4vHPV vaccine is recommended for use in males for prevention of persistent<br />

infection and anogenital disease caused by HPV types 6, 11, 16 and 18. <strong>The</strong><br />

4vHPV vaccine also provides protection against vaccine-type genital warts<br />

(which are mostly caused by HPV types 6 and 11). (See also 4.6.4 Vaccines above.)<br />

Children and adolescents aged 9–18 years<br />

<strong>The</strong> 4vHPV vaccine is recommended for males 9–18 years of age. <strong>The</strong> optimal<br />

age for administering the 4vHPV vaccine is approximately 11–13 years, as most<br />

males in this age group would not have commenced sexual activity and so<br />

would be naïve to all HPV types. Vaccination only provides protection against<br />

vaccine-type disease if the vaccine is delivered prior to acquisition of that HPV<br />

type. <strong>The</strong>refore, the decision to vaccinate older adolescent males, who may<br />

have already commenced sexual activities, should follow an assessment of the<br />

potential benefits, based on their likely previous HPV exposure and future risks<br />

of HPV exposure.<br />

Adults aged ≥19 years<br />

Vaccination of all men in this age group is not routinely recommended as many<br />

are likely to have been exposed to one or more vaccine HPV types through sexual<br />

activity (see 4.6.3 Epidemiology above).<br />

238 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


However, some adult males may gain an individual benefit from HPV<br />

vaccination. <strong>The</strong> decision to vaccinate older males should take into account their<br />

likelihood of previous exposure to HPV and their future risks of HPV exposure.<br />

Men who have sex with men<br />

<strong>The</strong> 4vHPV vaccine is recommended for men who have sex with men (MSM)<br />

who have not previously been vaccinated with 3 doses of HPV vaccine. <strong>The</strong><br />

decision to vaccinate males in this group should take into account their likelihood<br />

of previous exposure to HPV and their future risks of HPV exposure. Overall,<br />

MSM are at increased risk of persistent HPV infection and associated disease<br />

(independent of HIV status or the presence of other immunocompromising<br />

conditions). 14,38 In addition, at the population level, MSM are less likely to benefit<br />

from herd immunity attained from HPV vaccination of females. <strong>The</strong> safety and<br />

efficacy of 4vHPV vaccine has been demonstrated in MSM participants in a<br />

randomised clinical trial (see 4.6.4 Vaccines above).<br />

Persons who are immunocompromised<br />

HPV vaccine is recommended for adult men and women who are<br />

immunocompromised due to medical conditions (including HIV infection) or<br />

treatment. <strong>The</strong> decision to vaccinate immunocompromised persons should take<br />

into account their likelihood of previous exposure to HPV, their future risks<br />

of HPV exposure, and the extent and duration of their immunocompromise<br />

(see 3.3.3 Vaccination of immunocompromised persons). Immunocompromised<br />

adolescents who have not yet been vaccinated with 3 doses of HPV vaccine<br />

should be offered catch-up vaccination. This is based on evidence that persons<br />

who are immunocompromised are more likely to develop a persistent HPV<br />

infection and to subsequently progress to HPV-related disease. 14,91<br />

<strong>The</strong>re are currently no clinical trial data demonstrating the efficacy of either of<br />

the HPV vaccines in immunocompromised participants. However, 4vHPV has<br />

been shown to be well tolerated and immunogenic in HIV-infected males and<br />

women with systemic lupus erythematosus. 92-94 As HPV vaccines are not live<br />

viral vaccines, there are no specific safety concerns regarding administration<br />

to immunocompromised persons (see 3.3.3 Vaccination of immunocompromised<br />

persons).<br />

Cervical screening in vaccinated females<br />

For all sexually active women, regular cervical screening remains an important<br />

preventive measure against cervical disease (refer to the National Cervical<br />

Screening Program at www.cancerscreening.gov.au). Vaccination is not an<br />

alternative to cervical screening but is a complementary preventive measure, as<br />

HPV types other than those included in the current vaccines have the potential<br />

to cause cervical cancer. Likewise cervical screening is not an alternative to HPV<br />

vaccination. Both are recommended.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 239<br />

4.6 HUMAN PAPILLOMAVIRUS


Cervical screening detects histopathological changes. It is not recommended to<br />

test for the presence of HPV virus or antibody routinely as a way of determining<br />

whether HPV vaccination is indicated.<br />

For women who have recently been diagnosed with cervical dysplasia, or have<br />

been treated for this in the past, HPV vaccine will have no impact on current<br />

disease, but may prevent future dysplasia due to different HPV types included in<br />

the vaccine.<br />

4.6.8 Pregnancy and breastfeeding<br />

HPV vaccines are not recommended for pregnant women.<br />

Women who become pregnant after starting the HPV vaccination course should<br />

withhold getting further doses of the HPV vaccine while pregnant, and receive<br />

the remaining doses of the course after pregnancy.<br />

Females who inadvertently receive a dose of HPV vaccine around the time of<br />

conception or during pregnancy should be informed of the body of evidence<br />

supporting lack of harm from vaccine administration in this setting. Among<br />

women who became pregnant during the course of 4vHPV vaccine clinical trials<br />

(despite recommendations for participants to avoid pregnancy), the overall<br />

proportions of pregnancies that resulted in an adverse outcome (spontaneous<br />

abortion, late fetal death, infant with congenital anomalies) were similar among<br />

4vHPV vaccine recipients and placebo or control vaccine recipients. Although<br />

one clinical trial raised the possibility of an association between 4vHPV vaccine<br />

administered within 30 days following the estimated date of conception and an<br />

increased incidence of congenital anomalies in the infant, those conditions were<br />

relatively common and unrelated. 72 Pooled analyses from multiple clinical trials<br />

have not confirmed such an association. 95<br />

Pooled analysis of women who became pregnant during clinical trials showed<br />

that, overall, there were no differences in pregnancy outcomes between 2vHPV<br />

vaccine and control vaccine recipients. 96,97<br />

HPV vaccines can be given to breastfeeding women. Among breastfeeding<br />

mothers in the clinical studies of 4vHPV vaccine, the rates of adverse events<br />

in the mother and the breastfeeding infant were comparable between 4vHPV<br />

vaccine and control vaccination groups. 98 <strong>The</strong> effect on breastfed infants of<br />

the administration of 2vHPV vaccine to their mothers has not been evaluated<br />

directly in clinical studies, but breastfeeding is not considered a contraindication<br />

for receiving the 2vHPV vaccine.<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

240 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.6.9 Contraindications<br />

<strong>The</strong> only absolute contraindications to HPV vaccines are:<br />

• anaphylaxis following a previous dose of either HPV vaccine<br />

• anaphylaxis following any vaccine component.<br />

In particular, the 4vHPV vaccine is contraindicated in persons with a history of<br />

anaphylaxis to yeast.<br />

4.6.10 Adverse events<br />

Both the 2vHPV and 4vHPV vaccines are generally safe and well tolerated.<br />

For both vaccines, injection site pain was the most commonly reported adverse<br />

event (approximately 80% of recipients), followed by swelling and erythema<br />

(20–30% for each). Injection site reactions were more commonly reported in<br />

vaccine recipients than in recipients of aluminium-containing placebo or control<br />

vaccines in clinical trials. Systemic reactions were also very common following<br />

both vaccines, occurring in up to about 30% of recipients. <strong>The</strong> most common<br />

adverse events included headache, fatigue, fever and myalgia. In most of the<br />

clinical trials, the frequencies of most of these common systemic adverse events<br />

were comparable between the HPV vaccine and the control vaccine recipients.<br />

Meta-analyses on pooled data from multiple clinical trials on both the 2vHPV<br />

and 4vHPV vaccines have shown no increase in the risk of serious adverse events<br />

among vaccine recipients compared with control recipients. 99,100<br />

For both vaccines, the safety profile and the spectrum of adverse events<br />

following immunisation in males were similar to those reported in females of<br />

corresponding age groups, 74,75,101,102 although some of the studies were not direct<br />

comparison studies.<br />

Post-marketing passive surveillance of HPV vaccine use in the United States has<br />

identified syncope (fainting) as one of the most common adverse events reported<br />

following 4vHPV vaccine in adolescent and young adult females. 103 A small<br />

proportion (about 10%) of syncopal episodes resulted in a fall with head injury. 103<br />

Similar or higher rates of syncope have been reported in other countries, through<br />

different surveillance mechanisms. 104,105 However, a prospective adverse events<br />

surveillance study in the United States, based on over 600 000 records of vaccine<br />

doses administered, did not find any increased risk of syncope with 4vHPV<br />

vaccination compared to the expected rate following non-4vHPV vaccination in<br />

youths and adults. 106 Syncope (fainting) may follow any vaccination, especially<br />

in adolescents and young adults, but is preventable through appropriate<br />

precautions. (See also 2.3.2 Adverse events following immunisation). In an <strong>Australian</strong><br />

study, 22 subjects (including 14 with syncope and 8 with syncopal seizure<br />

following 4vHPV vaccination) were reviewed in a Victorian clinic and received<br />

further doses while supine; no recurrence of syncope occurred. 104<br />

Anaphylaxis and other suspected hypersensitivity reactions, including skin<br />

rash, after 4vHPV vaccine have also been reported. <strong>The</strong> estimated incidence<br />

PART 4 VACCINE-PREVENTABLE DISEASES 241<br />

4.6 HUMAN PAPILLOMAVIRUS


ate of anaphylaxis following 4vHPV vaccine in Australia, as at June 2010, was<br />

2.6 anaphylaxis episodes per million doses of vaccine distributed, which was<br />

within the rate range for other vaccines given to children and adolescents in<br />

international studies. 107 A prospective surveillance study in the United States<br />

did not find any increased risk of anaphylaxis or allergic reactions with 4vHPV<br />

vaccination compared to the expected rate following childhood vaccines. 106<br />

4.6.11 Variations from product information<br />

<strong>The</strong> product information for the 4vHPV vaccine, Gardasil, states that this<br />

vaccine is indicated for use in males up to 26 years of age and females up to<br />

45 years of age. <strong>The</strong> product information for the 2vHPV vaccine, Cervarix,<br />

states that this vaccine is indicated for use in females up to 45 years of<br />

age and is not registered for use in males of any age. <strong>The</strong> ATAGI instead<br />

recommends that some males aged >26 years, such as MSM and those who are<br />

immunocompromised, who are likely to derive an individual benefit from HPV<br />

vaccination, can be vaccinated with 4vHPV. <strong>The</strong> ATAGI also recommends that<br />

some females aged >45 years, such as those who are immunocompromised, can<br />

be vaccinated with either 2vHPV or 4vHPV, based on their individual risk of<br />

future HPV exposure and disease.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

242 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.7 INFLUENZA<br />

4.7.1 Virology<br />

<strong>The</strong> influenza viruses are single-stranded RNA orthomyxoviruses. <strong>The</strong>y are<br />

classified antigenically as types A, B or C, but only influenza A and B are<br />

clinically important in human disease. 1 Influenza viruses possess two surface<br />

glycoprotein antigens: the haemagglutinin (H), which is involved in cell<br />

attachment during infection, and the neuraminidase (N), which facilitates the<br />

release of newly synthesised virus from the cell. Influenza A viruses can be<br />

classified into subtypes based on differences in these surface antigens, whereas<br />

influenza B cannot. Antibody against the surface antigens, particularly the<br />

haemagglutinin, reduces infection or severe illness due to influenza.<br />

Both influenza A and influenza B viruses undergo frequent changes in their<br />

surface antigens, involving stepwise mutations of genes coding for H and N<br />

glycoproteins. This results in cumulative changes in influenza antigens, or<br />

‘antigenic drift’, which is responsible for the annual outbreaks and epidemics of<br />

influenza and is the reason that the composition of influenza vaccines requires<br />

annual review. ‘Antigenic shift’, defined as a dramatic change in influenza A<br />

H (and other) antigen, occurs occasionally and unpredictably and can cause<br />

pandemic influenza. 1 Pandemic subtypes arise following antigenic shift, which<br />

is due to direct adaptation to humans of an avian or animal virus, or to this<br />

adaptation occurring by genetic reassortment (mixing) with a human virus.<br />

4.7.2 Clinical features<br />

Influenza is transmitted from person to person by virus-containing respiratory<br />

aerosols produced during coughing or sneezing, or by direct contact with<br />

respiratory secretions. 1,2 Influenza virus infection causes a wide spectrum of<br />

disease, from no or minimal symptoms, to respiratory illness with systemic<br />

features, to multisystem complications and death from primary viral or<br />

secondary bacterial pneumonia. Severe disease is more likely with advanced<br />

age; lack of previous exposure to antigenically related influenza virus; greater<br />

virulence of the viral strain; chronic conditions, such as heart or lung disease,<br />

renal failure, diabetes and chronic neurological conditions; immunocompromise;<br />

pregnancy; and smoking. In pandemics, severe disease may also occur in<br />

otherwise healthy young adults. Annual attack rates in the general community<br />

are typically 5 to 10%, but may be up to 20% in some years. In households<br />

and ‘closed’ populations, attack rates may be 2 to 3 times higher. 3,4 However,<br />

as asymptomatic or mild influenza illness is common and symptoms are nonspecific,<br />

a large proportion of influenza infections are not detected.<br />

In adults, the onset of illness due to influenza is often abrupt, usually after an<br />

incubation period of 1 to 3 days, and includes systemic features such as malaise,<br />

feverishness, chills, headache, anorexia and myalgia. <strong>The</strong>se may be accompanied<br />

by a cough, nasal discharge and sneezing. Fever is a prominent sign of infection<br />

PART 4 VACCINE-PREVENTABLE DISEASES 243<br />

4.7 INFLUENZA


and peaks at the height of the systemic illness. Symptoms are similar for<br />

influenza A and B viruses. However, infections due to influenza A (H3N2)<br />

strains are more likely to lead to severe morbidity and increased mortality than<br />

influenza B or seasonal influenza A (H1N1) strains. 1,2<br />

<strong>The</strong> clinical features of influenza in infants and children are similar to those<br />

in adults. However, temperatures may be higher in children (and may result<br />

in febrile convulsions in this susceptible age group), and otitis media and<br />

gastrointestinal manifestations are more prominent. 5 Infection in young infants<br />

may be associated with more non-specific symptoms. 5,6<br />

Complications of influenza include: acute bronchitis, croup, acute otitis<br />

media, pneumonia (both primary viral and secondary bacterial pneumonia),<br />

cardiovascular complications including myocarditis and pericarditis,<br />

post-infectious encephalitis, Reye syndrome, and various haematological<br />

abnormalities. Primary viral pneumonia occurs rarely, but secondary bacterial<br />

pneumonia is a frequent complication in persons whose medical condition makes<br />

them vulnerable to the disease. Such persons are at high risk in epidemics and<br />

may die of pneumonia or cardiac decompensation.<br />

4.7.3 Epidemiology<br />

In most years, minor or major epidemics of type A or type B influenza occur,<br />

usually during the winter months in temperate regions. In Australia, although<br />

an average of 85 deaths and approximately 4000 hospitalisations directly<br />

attributed to influenza are notified annually, it has long been recognised that<br />

this is a substantial underestimate of the impact of influenza. 7 It is estimated that<br />

there are an average of over 13 500 hospitalisations due to influenza per year in<br />

Australia and over 3000 deaths per year in <strong>Australian</strong>s aged over 50 years alone. 8<br />

Influenza activity varies from year to year and is dependent on the circulating<br />

virus and the susceptibility of the population. 8,9 For example, there were over<br />

10 000 confirmed cases of influenza reported to the National Notifiable Diseases<br />

Surveillance System in the first half of 2011, compared with approximately 1570<br />

for the same period in 2010. 2 Laboratory testing for influenza has also increased<br />

in recent years. In Australia, like other developed countries, the highest influenza<br />

burden is seen in the elderly and in children


Figure 4.7.1: Influenza notification rates 2006–2007* and hospitalisation rates<br />

2005–2007,* Australia, by age group 9<br />

Rate per 100 000 population<br />

120<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

0–4<br />

5–9<br />

10–14<br />

15–19<br />

20–24<br />

Rate per 100 000 population<br />

25–29<br />

250<br />

200<br />

150<br />

100<br />

50<br />

30–34<br />

0<br />

35–39<br />

0 1 2<br />

Age (years)<br />

3 4<br />

40–44<br />

45–49<br />

50–54<br />

Age (years)<br />

55–59<br />

Notifications<br />

Hospitalisations<br />

* Notifications where the month of diagnosis was between January 2006 and December 2007;<br />

hospitalisations where the month of separation was between July 2005 and 30 June 2007. <strong>The</strong>se<br />

data are prior to the appearance of the pandemic A(H1N1)pdm09 virus in 2009.<br />

Three pandemics were recognised in the 20th century, in 1918 (H1N1),<br />

1957 (H2N2) and 1968 (H3N2). Each of these pandemic strains replaced the<br />

previously circulating influenza A subtype and went on to circulate as seasonal<br />

influenza. In 1977, the A (H1N1) re-emerged in the human population and, since<br />

then, A (H1N1) and A (H3N2) have co-circulated with influenza B. Recently,<br />

the avian influenza A virus subtypes, H5N1 and H9N2, have caused human<br />

infections. <strong>The</strong> most notable of these is the A (H5N1) subtype, which has become<br />

established in domestic poultry throughout Southeast Asia and has spread to<br />

Europe and Africa in either wild birds or domestic poultry. Although growing<br />

numbers of people have contracted the virus by contact with birds and there is<br />

a high mortality rate (≥50%), there has been no evidence of ongoing person-toperson<br />

transmission.<br />

In 2009, the World Health Organization (WHO) declared a pandemic of a novel<br />

subtype A (H1N1) influenza virus, A(H1N1)pdm09, which originated in swine;<br />

the pandemic started in Mexico and the United States in April 2009. In August<br />

2010, the WHO reported that more than 214 countries and overseas territories or<br />

60–64<br />

65–69<br />

70–74<br />

75–79<br />

80–84<br />

85<br />

PART 4 VACCINE-PREVENTABLE DISEASES 245<br />

4.7 INFLUENZA


communities reported laboratory-confirmed cases of A(H1N1)pdm09 (pH1N1)<br />

influenza, including more than 18 000 deaths. 14 In Australia, a total of 44 403<br />

confirmed cases of pH1N1 influenza occurred between May 2009 and November<br />

2010, including 6767 cases in 2010. A total of 213 pandemic influenza-associated<br />

deaths were reported, 22 of which occurred in 2010. 15<br />

Evidence from multiple outbreak sites demonstrated that the A(H1N1)pdm09<br />

virus rapidly established itself and was the dominant influenza strain in most<br />

parts of the world. <strong>The</strong> clinical picture of pH1N1 influenza appeared to be largely<br />

consistent across all countries, with most affected persons experiencing moderate<br />

illness. Risk factors for severe disease included obesity, pregnancy, diabetes<br />

mellitus and, in Australia, being of Aboriginal or Torres Strait Islander descent<br />

(see 3.1 Vaccination for Aboriginal and Torres Strait Islander people). Although<br />

influenza can cause very severe and fatal illness, particularly in the elderly, the<br />

impact of pH1N1 influenza in younger healthy adults and in pregnant women<br />

was proportionally greater than normal seasonal outbreaks, even though the<br />

absolute number of such cases remained low. During the last quarter of 2009,<br />

Australia introduced a non-adjuvanted, inactivated, egg-derived, monovalent<br />

A(H1N1)pdm09 vaccine for administration to all persons aged ≥6 months<br />

(Panvax, CSL Limited). Expired monovalent vaccine supplies were withdrawn<br />

in December 2010. 16 In Australia, there was ongoing summer activity of A(H1N1)<br />

pdm09 in late 2009, and the virus continued to circulate in 2010 and 2011,<br />

replacing the previously circulating seasonal H1N1 strain. 2 <strong>The</strong> A(H1N1)pdm09<br />

strain was included in trivalent seasonal influenza vaccine formulations used in<br />

the southern hemisphere in 2010, 2011 and 2012.<br />

4.7.4 Vaccines<br />

<strong>The</strong> administration of influenza vaccine to persons at risk of complications of<br />

infection is the single most important measure in preventing or attenuating<br />

influenza infection and preventing mortality. After vaccination, most adults<br />

develop antibody levels that are likely to protect them against the strains of<br />

virus represented in the vaccine. In addition, there is likely to be protection<br />

against many related influenza variants. Infants, the very elderly, and persons<br />

who are immunocompromised may develop lower post-vaccination antibody<br />

levels. Under these circumstances, influenza vaccine may be more effective<br />

in preventing lower respiratory tract involvement or other complications of<br />

influenza than in preventing influenza infection.<br />

246 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Always check annual seasonal influenza vaccine availability statements<br />

on www.immunise.health.gov.au. Vaccines and age eligibility change<br />

from year to year.<br />

Vaccines for intramuscular administration<br />

Children aged ≥6 months and adults (see Table 4.7.1 for dosage information for<br />

different age groups)<br />

• Agrippal – Novartis Vaccines and Diagnostics Pty Ltd (inactivated<br />

influenza virus). Each 0.5 mL pre-filled syringe contains 15 µg<br />

haemagglutinin of each of the three recommended strains. May<br />

contain traces of kanamycin, neomycin, formaldehyde, barium<br />

sulphate, cetrimonium bromide (CTAB), polysorbate 80 and egg<br />

protein.<br />

• Fluarix – GlaxoSmithKline (inactivated influenza virus). Each 0.5 mL<br />

pre-filled syringe contains 15 µg haemagglutinin of each of the<br />

three recommended strains. May contain traces of formaldehyde,<br />

gentamicin, polysorbate 80, octoxinol 10 and egg protein.<br />

• Influvac – Abbott Products Pty Ltd (inactivated influenza virus). Each<br />

0.5 mL pre-filled syringe contains 15 µg haemagglutinin of each of<br />

the three recommended strains. May contain traces of formaldehyde,<br />

CTAB, polysorbate 80, gentamicin and egg protein.<br />

• Influvac Junior (6 months to


Children aged ≥10 years* and adults (see Table 4.7.1 for dosage information)<br />

• Fluvax – CSL Limited (inactivated influenza virus).* Each 0.5 mL<br />

pre-filled syringe contains 15 µg haemagglutinin of each of the three<br />

recommended strains. May contain traces of neomycin, polymyxin B,<br />

β-propiolactone, sodium taurodeoxycholate and egg protein.<br />

Adults aged ≥65 years<br />

• Fluad – Novartis Vaccines and Diagnostics Pty Ltd (inactivated<br />

influenza virus). Each 0.5 mL pre-filled syringe contains 15 µg<br />

haemagglutinin of each of the three recommended strains, adjuvanted<br />

with MF59C.1 (which contains squalene and polysorbate 80). May<br />

contain traces of kanamycin, neomycin, formaldehyde, barium<br />

sulphate, CTAB and egg protein.<br />

Vaccines for intradermal administration<br />

Adults aged 18–59 years<br />

• Intanza 9 μg – Sanofi Pasteur Pty Ltd (inactivated influenza virus).<br />

Each 0.1 mL pre-filled purpose-designed Micro-Injection System<br />

contains 9 µg haemagglutinin of each of the three recommended<br />

strains. May contain traces of formaldehyde, octoxinol 9, neomycin<br />

and egg protein.<br />

Adults aged ≥60 years<br />

• Intanza 15 μg – Sanofi Pasteur Pty Ltd (inactivated influenza virus).<br />

Each 0.1 mL pre-filled purpose-designed Micro-Injection System<br />

contains 15 µg haemagglutinin of each of the three recommended<br />

strains. May contain traces of formaldehyde, octoxinol 9, neomycin<br />

and egg protein.<br />

* Fluvax (CSL Limited) is registered by the <strong>The</strong>rapeutic Goods Administration (TGA) for<br />

administration in children ≥5 years of age; however, it is not recommended for use in children<br />


7.5 µg of viral haemagglutinin (in a 0.25 mL dose) of each of the same three<br />

strains found in the adult formulations.<br />

All the influenza vaccines currently available in Australia are either split virion<br />

or subunit vaccines prepared from purified inactivated influenza virus that has<br />

been cultivated in embryonated hens’ eggs. Split virion and subunit vaccines<br />

are generally considered to be equivalent with respect to safety, and both<br />

are substantially free of the systemic reactions sometimes induced by whole<br />

virus vaccines. One exception is Fluvax (CSL Limited), which in 2010 resulted<br />

in higher rates of adverse events, specifically fevers and febrile convulsions,<br />

in children aged


efficacy varied by influenza season. 29 <strong>The</strong> efficacy of inactivated influenza vaccine<br />

against influenza-like illness in persons ≥65 years of age living in the community<br />

is estimated to be 43% when viral circulation is high, although there have been<br />

few randomised controlled trials of influenza vaccine in elderly people. 30 A<br />

systematic review assessing vaccine effectiveness estimated that during periods<br />

of high virus circulation, when vaccine match is good, influenza vaccination<br />

is approximately 45% effective against hospitalisations due to influenza and<br />

pneumonia and 60% effective against all-cause mortality in persons aged >65<br />

years in nursing home settings. 30<br />

Currently available influenza vaccines confer protection for about a year. Low<br />

levels of protection may persist for a further year, if the prevalent strain remains<br />

the same or undergoes only minor antigenic drift. 13,27 Continuing protection<br />

requires annual vaccination with vaccine containing the most recent strains.<br />

4.7.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 31 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

At the end of each year, influenza vaccines should be appropriately discarded<br />

to avoid inadvertently using a product with incorrect formulation in the<br />

following year.<br />

4.7.6 Dosage and administration<br />

Vaccines registered by the TGA, and the ages for which they are indicated,<br />

can change from year to year. Always check annual seasonal influenza vaccine<br />

availability statements on www.immunise.health.gov.au, and consult individual<br />

product information.<br />

Influenza vaccines available in Australia for IM use are presented in pre-filled<br />

syringes, of either 0.5 mL or 0.25 mL. <strong>The</strong> dose of vaccine to be administered<br />

varies by age, with children aged 6 months to


Table 4.7.1: Recommended doses of influenza vaccine<br />

Age Dose Number of doses (first<br />

year of vaccination)<br />

Number of doses*<br />

(subsequent years)<br />

6 months to


All adults aged ≥65 years<br />

<strong>The</strong>re is evidence that influenza vaccine reduces hospitalisations from influenza<br />

and pneumonia and all-cause mortality in adults ≥65 years of age. 30 (See also<br />

4.7.4 Vaccines above.)<br />

Persons at increased risk of complications from influenza infection<br />

Persons aged ≥6 months with conditions predisposing to severe influenza, 7<br />

such as:<br />

• Pregnancy – Pregnant women (and women planning pregnancy) are<br />

recommended to be immunised against influenza because they are at<br />

increased risk of morbidity and mortality from influenza and because<br />

there is good evidence that influenza immunisation in pregnancy is safe<br />

and effective. 38-45 <strong>The</strong> risk to the mother of complications from influenza<br />

increases in the later stages of pregnancy. 38-42,46-50 <strong>The</strong>re is also a growing<br />

body of evidence showing that influenza vaccination of pregnant women<br />

protects infants against influenza for the first 6 months after birth. 39,51,52 Most<br />

evidence around infant protection is from studies of maternal influenza<br />

vaccination in the second or third trimester. 38-42,46-51 Influenza vaccination is<br />

thought to provide protection for up to a year, but there is limited evidence<br />

that immunity may start to wane from 4 months following immunisation.<br />

Although it is recommended that all pregnant women should be immunised<br />

as early as possible in pregnancy, 53 the precise timing of vaccination will<br />

depend on the time of the year, vaccine availability, influenza seasonality,<br />

gestation of pregnancy and the likely duration of immunity. (See also 4.7.8<br />

Pregnancy and breastfeeding below.)<br />

• Cardiac disease, including cyanotic congenital heart disease, coronary artery<br />

disease and congestive heart failure – Influenza causes increased morbidity<br />

and mortality in children with congenital heart disease and adults with<br />

coronary artery disease and congestive heart failure. 37,54-57<br />

• Down syndrome – Persons with Down syndrome should receive annual<br />

seasonal influenza vaccine whether or not they have congenital heart<br />

disease. This is due to the presence of anatomical abnormalities, which put<br />

them at increased risk of upper respiratory tract infections, as well as a high<br />

prevalence of other medical conditions that put them at increased risk of<br />

severe influenza. 56<br />

• Obesity – Persons with significant obesity, defined as a BMI ≥30 kg/m2 , with<br />

or without other underlying conditions, have been identified as being at<br />

increased risk for hospitalisation with respiratory complications following<br />

influenza. 37 This risk was particularly apparent during the 2009–2010<br />

pandemic. 58,59<br />

252 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


• Chronic respiratory conditions, including:<br />

» Suppurative lung disease, bronchiectasis and cystic fibrosis37,55 – Patients<br />

with these diseases are at greatly increased risk from influenza, which may<br />

cause irreversible deterioration in lung function. 60<br />

» Chronic obstructive pulmonary disease (COPD) and chronic<br />

emphysema37,61,62 – Data from several studies provide evidence that<br />

influenza vaccination has a clinically important protective effect on<br />

influenza-related COPD exacerbations, and probably an effect on the total<br />

number of exacerbations in COPD patients. 61,62<br />

» Severe asthma – In patients with severe asthma, defined as requiring<br />

frequent hospital visits and the use of multiple medications, annual<br />

influenza vaccine is an important part of routine care. 37,55 <strong>The</strong>re are<br />

insufficient data from randomised controlled trials of influenza vaccine<br />

to define efficacy across the whole spectrum of asthma, but influenza can<br />

cause severe exacerbations of wheezing, and about 10% of episodes of<br />

virus-induced wheezing are attributable to influenza.<br />

• Chronic neurological conditions (e.g. multiple sclerosis, spinal cord injuries,<br />

seizure disorders or other neuromuscular disorders) – <strong>The</strong>se conditions<br />

can compromise respiratory function or the expulsion of respiratory<br />

secretions that can then increase the risk for aspiration. 37,55 Influenza<br />

vaccination is particularly important for children ≥6 months of age with<br />

chronic neurological conditions as these children can experience severe,<br />

even fatal, influenza.<br />

• Immunocompromising conditions – Persons who are immunocompromised,<br />

including those with HIV infection, malignancy or chronic steroid<br />

use, are at an increased risk from influenza (see 3.3,3 Vaccination of<br />

immunocompromised persons). 37,55 <strong>The</strong>y may also have a reduced immune<br />

response to the vaccine, although influenza vaccination affords some<br />

protection. 63 Influenza vaccination is recommended annually in all oncology<br />

patients aged ≥6 months.<br />

All immunocompromised persons, irrespective of age, who receive influenza<br />

vaccine for the first time, are recommended to receive 2 vaccine doses, at<br />

least 4 weeks apart, and 1 dose annually thereafter. Where it is known that<br />

a new influenza vaccine strain is circulating in the community to which<br />

cross-protective immunity in the population is low (such as in the setting of<br />

an influenza pandemic), it may be appropriate that immunocompromised<br />

persons receive 2 doses of inactivated influenza vaccine, a minimum of<br />

4 weeks apart, to achieve an optimal immune response. For example,<br />

in the 2009–2010 H1N1 global influenza pandemic it was shown that<br />

seroconversion to influenza vaccination in immunocompromised adolescents<br />

and adults was improved following receipt of 2 vaccine doses. 64 Further<br />

PART 4 VACCINE-PREVENTABLE DISEASES 253<br />

4.7 INFLUENZA


information and annual influenza vaccine recommendations are available on<br />

the Immunise Australia website (www.imunise.health.gov.au).<br />

While patients with advanced HIV disease and low CD4 + T-lymphocyte<br />

counts may not develop protective antibody titres, there is evidence that<br />

for those with minimal symptoms and high CD4 + T-lymphocyte counts,<br />

protective antibody titres are obtained after influenza vaccination. Influenza<br />

vaccine has been shown to reduce the incidence of influenza in HIVinfected<br />

patients, and, although viral load may increase transiently, there<br />

was no impact on CD4 + count. 63 (See also 3.3 Groups with special vaccination<br />

requirements, Table 3.3.4.)<br />

• Other chronic illnesses requiring regular medical follow-up or hospitalisation<br />

in the preceding year, including:<br />

» diabetes mellitus37,55,65 » chronic renal failure37,55 » alcoholism66,67 » haemoglobinopathies55 » chronic inherited metabolic diseases (which includes amino acid disorders,<br />

carbohydrate disorders, cholesterol biosynthesis disorders, fatty acid<br />

oxidation defects, lactic acidosis, mitochondrial disorders, organic acid<br />

disorders, urea cycle disorders, vitamin/cofactor disorders, porphyrias) 37,55<br />

• Long-term aspirin therapy in children (aged 6 months to 10 years) – Such<br />

children are at increased risk of Reye syndrome after influenza. 68,69<br />

Aboriginal and Torres Strait Islander people aged ≥15 years<br />

Annual influenza vaccination is recommended for all Aboriginal and Torres<br />

Strait Islander people ≥15 years of age, in view of their substantially increased<br />

risk of hospitalisation and death from influenza and pneumonia. 70 (See also 3.1<br />

Vaccination for Aboriginal and Torres Strait Islander people and 4.13 Pneumococcal<br />

disease.)<br />

Children aged


Residents of residential aged care facilities and long-term residential facilities<br />

Annual influenza vaccination is recommended for residents of these facilities,<br />

including inmates of correctional facilities, due to high rates of influenza<br />

transmission and complications during outbreaks in such facilities. 12,13,62<br />

Homeless people<br />

<strong>The</strong> living conditions and prevalence of underlying medical conditions among<br />

homeless people will predispose them to complications and transmission of<br />

influenza.<br />

Persons who may transmit influenza to persons at increased risk of<br />

complications from influenza infection<br />

<strong>The</strong> following groups of people can potentially transmit influenza to persons<br />

at increased risk of complications from influenza infection; vaccination of these<br />

groups is therefore recommended to protect those at risk:<br />

• all healthcare providers (particularly those of immunocompromised patients)<br />

• staff (or volunteers) working in nursing homes<br />

• staff (or volunteers) working in long-term care facilities<br />

• household contacts (including children ≥6 months of age) of those in highrisk<br />

groups, including providers of home care to persons at risk of high<br />

influenza morbidity<br />

• staff working in early childhood education and care<br />

• staff (or volunteers) providing care to homeless people.<br />

Persons involved in the commercial poultry or pork industry or in culling<br />

poultry or pigs during confirmed avian or swine influenza activity<br />

Vaccination using the seasonal influenza vaccine composition current at the<br />

time is recommended for poultry or piggery workers and others in regular close<br />

contact with poultry or pigs during an avian or swine influenza outbreak. 75<br />

Although routine seasonal influenza vaccine does not protect against avian or<br />

swine influenza, there is a possibility that a person who is infected at the same<br />

time with animal and human strains of influenza virus could act as a vessel for<br />

reassortment of the two strains to form a virulent strain, with the potential for<br />

spread from human to human (i.e. initiate a pandemic as was the case with swine<br />

influenza in 2009). 76 In addition, vaccination can also prevent the transmission of<br />

influenza from humans to animals.<br />

Persons providing essential services<br />

Vaccination of those who provide essential community services will minimise<br />

disruption of essential activities during influenza outbreaks. Influenza viral<br />

infections can place considerable pressure upon both public and private<br />

healthcare services (see 3.3 Groups with special vaccination requirements, Table<br />

3.3.7 Recommended vaccinations for persons at increased risk of certain occupationally<br />

acquired vaccine-preventable diseases).<br />

PART 4 VACCINE-PREVENTABLE DISEASES 255<br />

4.7 INFLUENZA


Workers in other industries<br />

Due to the high attack rate of influenza in the general population, influenza<br />

vaccination in the workplace can result in benefits such as increased productivity<br />

and reduced absenteeism among workers. 77 Employers should consider the<br />

benefits of offering influenza vaccine in their individual workplace.<br />

Travellers<br />

Influenza vaccine is particularly relevant if influenza epidemics are occurring at<br />

the traveller’s destination(s). Travellers in large tourist groups, especially those<br />

including older people, those travelling on cruises, and/or those who are likely<br />

to be in confined circumstances for days to weeks, are at risk of influenza, either<br />

acquired before departure or from travel to areas of the world where influenza<br />

is currently circulating. Influenza vaccination is recommended if travelling<br />

during the influenza season, especially if it is known before travel that influenza<br />

is circulating in the destination region. 78 (See also 3.2 Vaccination for international<br />

travel.)<br />

4.7.8 Pregnancy and breastfeeding<br />

Influenza vaccination is recommended for pregnant women (see 4.7.7<br />

Recommendations above) and is safe to administer during any stage of pregnancy<br />

or while breastfeeding. 44,79-81<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.7.9 Contraindications<br />

<strong>The</strong> only absolute contraindications to influenza vaccines are:<br />

• anaphylaxis following a previous dose of any influenza vaccine<br />

• anaphylaxis following any vaccine component.<br />

See 4.7.10 Precautions below for persons with a known egg allergy.<br />

4.7.10 Precautions<br />

Persons with known egg allergy82-86 Several recently published reviews, guidelines and reports have indicated that<br />

the risk of anaphylaxis associated with influenza vaccination of egg-allergic<br />

patients is very low. 82-86 Persons with egg allergy, including anaphylaxis, can be<br />

safely vaccinated with influenza vaccines that have less than 1 μg of residual egg<br />

ovalbumin per dose. 82,84 Due to changes in influenza vaccine manufacturing, the<br />

majority of influenza vaccines currently used contain less than 1 μg of ovalbumin<br />

per dose. 82 Note the amount of residual egg ovalbumin may vary from year to<br />

year due to manufacturing processes, batches and country of origin. 82,84,85 <strong>The</strong><br />

product information (PI) of the vaccine to be given should be checked for the<br />

vaccine’s ovalbumin content prior to vaccine administration. Allergy testing with<br />

256 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


influenza vaccine prior to administration is not recommended as there is poor<br />

correlation between test results and vaccine tolerance. 82,84-86<br />

However, there is still a low risk of anaphylaxis, so it is essential that such<br />

patients are vaccinated in facilities with staff able to recognise and treat<br />

anaphylaxis. Additional information on influenza vaccination of individuals<br />

with an allergy to eggs, including risk, dosage and observation period, can be<br />

found in the Australasian Society of Clinical Immunology and Allergy (ASCIA)<br />

guidelines. 84 (See also ‘Vaccination of persons with a known egg allergy’ in 3.3.1<br />

Vaccination of persons who have had an adverse event following immunisation.)<br />

Persons with a history of Guillain-Barré syndrome<br />

Persons with a history of Guillain-Barré syndrome (GBS) have an increased<br />

likelihood in general of developing GBS again, and the chance of them<br />

coincidentally developing the syndrome following influenza vaccination may be<br />

higher than in persons with no history of GBS. 87 Diagnosis of GBS is complex and<br />

must be made by a physician. (See also 4.7.11 Adverse events below.)<br />

Children requiring both influenza and 13-valent pneumococcal<br />

conjugate vaccine<br />

Parents/carers of infants or children who are recommended to receive both<br />

influenza vaccine and 13vPCV should be advised of the slightly higher risk<br />

of fever following concomitant administration of these vaccines (see 4.13<br />

Pneumococcal disease). Given this relatively low increase in risk, providing<br />

13vPCV and inactivated trivalent influenza vaccine concurrently to children aged<br />

12–23 months is acceptable; however, immunisation service providers should<br />

advise parents regarding this, and provide the option of administering these two<br />

vaccines on separate days (with an interval of not less than 3 days). In the event<br />

that fever occurs following either vaccine, an interval may minimise the risk of<br />

increased adverse reactions.<br />

4.7.11 Adverse events<br />

Fever, malaise and myalgia occur commonly, in 1 to 10% of persons who receive<br />

influenza vaccination. 88-90 <strong>The</strong>se adverse events may commence within a few<br />

hours of vaccination and may last for 1 to 2 days. 88-90 In children


Local adverse events (induration, redness, swelling and pruritus) occur more<br />

commonly following intradermal influenza vaccine administration. However,<br />

most patients report that these events are transient and resolve fully within a<br />

few days. 93<br />

Post-vaccination symptoms may mimic influenza infection, but all currently<br />

available influenza vaccines do not contain live virus and so do not cause<br />

influenza.<br />

Immediate adverse events (such as hives, angioedema or anaphylaxis) are a<br />

rare consequence of influenza vaccination. <strong>The</strong>y probably represent an allergic<br />

response to a residual component of the manufacturing process, most likely egg<br />

protein. 82,84 Persons with a history of anaphylaxis after eating eggs or a history<br />

of a severe allergic reaction following occupational exposure to egg protein may<br />

receive influenza vaccination after medical consultation. 82,84<br />

A small increased risk of GBS was associated historically with one influenza<br />

vaccine in the United States in 1976, but, since then, close surveillance has shown<br />

that GBS has occurred at a very low rate of up to 1 in 1 million doses of influenza<br />

vaccine, if at all. 94 Diagnosis of GBS is complex and must be made by a physician<br />

(see ‘Uncommon/rare AEFI’ in 2.3.2 Adverse events following immunisation).<br />

Narcolepsy (sudden sleeping illness) has been described predominantly in the<br />

Scandinavian population, in association with adjuvanted pandemic influenza<br />

vaccines. 95,96 <strong>The</strong>se vaccines were not used and are not available in Australia.<br />

4.7.12 Public health management of influenza<br />

Laboratory-confirmed cases of influenza are notifiable in all states and territories<br />

in Australia. Detailed information regarding the management of influenza cases<br />

and contacts can be found in the national guidelines for control of influenza97 (www.health.gov.au/cdnasongs).<br />

Further instructions about the public health management of influenza can also<br />

be obtained from state/territory public health authorities (see Appendix 1<br />

Contact details for <strong>Australian</strong>, state and territory government health authorities and<br />

communicable disease control).<br />

4.7.13 Variations from product information<br />

<strong>The</strong> product information for influenza vaccines lists allergy to eggs, chicken<br />

feathers and/or some food proteins as a contraindication. <strong>The</strong> ATAGI<br />

recommends instead that patients with egg allergies and other allergies can<br />

be vaccinated under strict medical supervision, in accordance with ASCIA<br />

guidelines. 84<br />

<strong>The</strong> product information for Fluvax (CSL Limited) states that this vaccine is<br />

registered for use in children ≥5 years of age. However, the ATAGI does not<br />

recommend the use of this vaccine in children aged


4.8 JAPANESE ENCEPHALITIS<br />

4.8.1 Virology<br />

Japanese encephalitis (JE) is caused by a mosquito-borne RNA flavivirus.<br />

4.8.2 Clinical features<br />

<strong>The</strong> disease is typically an acute neurological illness, characterised by headache,<br />

fever, convulsions, focal neurological signs and depressed level of consciousness.<br />

It has a high case-fatality rate and there is a high prevalence of neurological<br />

sequelae (up to 50%) in those who survive the acute illness. 1 Less commonly, the<br />

disease may present as an acute flaccid paralysis. 1 Milder forms include febrile<br />

illness with headache, and aseptic meningitis. It is recognised, however, that<br />

most infections are asymptomatic; published estimates of the symptomatic to<br />

asymptomatic infection ratio vary in different populations from 1:25 to 1:1000. 1<br />

4.8.3 Epidemiology<br />

JE is a significant public health problem in many parts of Asia, including the<br />

Indian subcontinent, Southeast Asia and China. 1 In recent years, however, the<br />

disease has extended beyond its traditionally recognised boundaries with, for<br />

example, occasional cases in eastern Indonesia, occasional outbreaks in the<br />

Torres Strait and 1 case in north Queensland. 2,3 JE is now considered endemic<br />

in the Torres Strait region and Papua New Guinea. 2-5 <strong>The</strong> first ever outbreak of<br />

Japanese encephalitis (JE) in Australia occurred in the remote outer islands of the<br />

Torres Strait in 1995, with 3 cases, 2 of them fatal. <strong>The</strong>re have been 5 cases to date<br />

acquired in Australia. JE virus has only been detected infrequently in sentinel<br />

animal surveillance in the outer islands. <strong>The</strong> sentinel pig surveillance system<br />

has been gradually disbanded since 2006, with surveillance of the last remaining<br />

herd on Cape York ceasing from the 2011–2012 wet season.<br />

<strong>The</strong>re has not been a case of JE in the Torres Strait since 1998 and the risk of<br />

JE has diminished considerably in the outer islands since the mid-1990s. Most<br />

communities have relocated pigs well away from homes, and major drainage<br />

works on most islands have markedly reduced potential breeding sites for vector<br />

mosquitoes. Between 2001 and April 2012, only 4 cases of JE virus infection were<br />

notified in Australia. 6<br />

<strong>The</strong> JE virus is essentially a zoonosis of pigs and wading birds, and is transmitted<br />

between these animals by culicine mosquitoes. 1 <strong>The</strong> virus replicates, leading to a<br />

transient high-level viraemia, within these so-called ‘amplifying’ hosts, but not<br />

within other large vertebrates such as horses and humans.<br />

Indeed, humans are an incidental host, infected when living in close proximity<br />

to the enzootic cycle; this usually occurs in rural areas where there is prolific<br />

breeding of the vectors in flooded rice fields. 1<br />

<strong>The</strong>re are two recognised epidemiological patterns of JE. 1 In the temperate or<br />

subtropical regions of Asia (northern Thailand, northern Vietnam, Korea, Japan,<br />

PART 4 VACCINE-PREVENTABLE DISEASES 259<br />

4.8 JAPANESE ENCEPHALITIS


Taiwan, China, Nepal and northern India), the disease occurs in epidemics<br />

during the summer or wet season months (April to May until September to<br />

October). In the tropical regions (most of Southeast Asia, Sri Lanka, southern<br />

India), the disease is endemic, occurring throughout the year, but particularly<br />

during the wet season. 1 A 2006 study confirmed that JE virus is hyperendemic in<br />

Bali, that it causes substantial human illness, and that it circulates year round. 7<br />

In some countries (Japan, Taiwan, South Korea and some provinces of China),<br />

the incidence of JE has declined considerably in recent decades, and it has been<br />

eradicated from Singapore. <strong>Immunisation</strong>, changes in pig husbandry, a reduction<br />

in land utilised for rice farming, and improved socioeconomic circumstances<br />

have all contributed to these changes. 1 Updated information regarding JE virus<br />

activity and/or risk in travel destinations should be sought from a reputable<br />

source prior to travel. 8<br />

4.8.4 Vaccines<br />

• Imojev – Sanofi Pasteur Pty Ltd (live attenuated Japanese encephalitis<br />

virus). Lyophilised powder in a monodose vial with separate diluent.<br />

Each 0.5 mL reconstituted dose contains 4.0–5.8 log plaque-forming<br />

units (PFU) of live attenuated Japanese encephalitis virus; mannitol;<br />

lactose; glutamic acid; potassium hydroxide; histidine; human serum<br />

albumin. No adjuvants or antibiotics are added.<br />

• JEspect – Intercell Biomedical Ltd/CSL Limited (inactivated Japanese<br />

encephalitis virus). Each 0.5 mL pre-filled syringe contains 6 μg of<br />

purified inactivated Japanese encephalitis virus; 0.25 mg aluminium<br />

as aluminium hydroxide. No preservatives or antibiotics are added.<br />

Two JE vaccines, each with different characteristics, are available for use<br />

in Australia. <strong>The</strong> inactivated mouse brain-derived JE vaccine formulation<br />

manufactured in Japan, JE-Vax, which was previously used in Australia, is no<br />

longer manufactured. Clinical and animal studies have provided evidence in<br />

support of an immunological correlate of immunity (established by the World<br />

Health Organization as a neutralising antibody titre of ≥1:10). Both currently<br />

available JE vaccines were registered on the basis of this serological correlate in<br />

lieu of a field efficacy trial.<br />

Imojev is a live attenuated, monovalent viral vaccine produced using<br />

recombinant technology. Two genes of the 17D-204 yellow fever vaccine virus<br />

have been replaced with two genes, prM and E genes, from the Japanese<br />

encephalitis virus strain SA 14-14-2. About 94% of healthy adults aged<br />

18–84 years seroconverted to a strain homologous to that in the Imojev vaccine<br />

14 days after a single vaccine dose. 9 Several clinical trials have demonstrated that,<br />

260 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


28 days following vaccination with a single dose of Imojev, protective levels of<br />

neutralising antibodies against the homologous vaccine virus strain are present<br />

in 96% of vaccine-naïve children aged 12–24 months10 and 99% of adults. 9-12<br />

Immunogenicity was non-inferior to that attained after a 3-dose primary course<br />

of the inactivated mouse brain-derived JE vaccine9 that was previously used in<br />

Australia. Subjects also seroconverted to various wild-type, non-homologous,<br />

JE virus strains (70 to 97% of children aged 12–24 months and 70 to 100% of<br />

adults); 10,13 85% of adults developed neutralising antibodies against all four<br />

wild-type strains used for testing. 12 Protective antibody to the vaccine strain was<br />

maintained at 6 months after vaccination in 87% of children aged 12–24 months, 10<br />

and 97% of adults. 12 About 93% of adults maintained a protective antibody level<br />

to the vaccine strain, and 65% to at least three wild-type strains, 60 months after<br />

a single vaccine dose. 12 Establishment of immunological memory in vaccinated<br />

adult subjects has also been demonstrated. 13<br />

Among children aged 2–5 years previously vaccinated with a mouse brainderived<br />

JE vaccine (of whom 86% were seropositive against the vaccine strain<br />

and 72 to 81% seropositive against four wild-type strains at baseline), a dose<br />

of Imojev produced seroprotective antibody levels against the vaccine strain<br />

in 100%, and against all wild-type strains in 99%. Ninety-three per cent of the<br />

children seroconverted to the vaccine strain and 90% against all wild-type<br />

strains. 10<br />

JEspect is a Vero cell-derived, inactivated, aluminium-adjuvanted vaccine<br />

based on the attenuated SA 14-14-2 JE virus strain. JEspect has equivalent<br />

immunogenicity (after 2 doses, given 4 weeks apart) to 3 doses of the previously<br />

available mouse brain-derived vaccine, with seroconversion achieved in 98% of<br />

subjects. 14 Post-vaccination geometric mean titres (GMT) in JEspect recipients<br />

were significantly higher than GMTs attained after a mouse brain-derived<br />

vaccine. 14 After a standard 2-dose course, protective levels of neutralising<br />

antibodies have been found to persist for 6 months in 95%, for 12 months in 83%<br />

and for 24 months in 48% of JEspect-vaccinated subjects in central Europe, 15,16<br />

but in 83%, 58% and 48%, respectively, at these three time points in subjects<br />

in western and northern Europe. 17 A suggested plausible explanation for the<br />

discrepancy between these two studies is prior vaccination with the tick-borne<br />

encephalitis vaccine in a large proportion of subjects in the central European<br />

study. 16,17 In an extension of the western and northern European study, those who<br />

did not have a seroprotective antibody level at either the 6- or 12-month followup<br />

point were given a booster dose at 11 and/or 23 months after first vaccination;<br />

seropositivity was attained in 100% of these subjects. 17 Another study showed<br />

that a booster dose given 15 months after the primary immunisation with 2<br />

doses of JEspect increased the GMT by 5-fold after 4 weeks, and the proportion<br />

of subjects with seroprotective antibody levels increased from 69.2% pre booster<br />

to 98.5% at 6 and 12 months post booster. Mathematical modelling has predicted<br />

PART 4 VACCINE-PREVENTABLE DISEASES 261<br />

4.8 JAPANESE ENCEPHALITIS


that 95% of subjects would maintain seroprotective antibodies for 3.8 years after<br />

a booster dose. 18<br />

A phase II study in 60 Indian children aged 1 to


When using JEspect in children aged ≥3 years and adults, primary vaccination<br />

consists of 2 doses, each of 0.5 mL, according to the following schedule:<br />

• 1st dose at day 0<br />

• 2nd dose 28 days after the 1st dose.<br />

Do not mix either Japanese encephalitis vaccine with any other vaccine in the<br />

same syringe.<br />

Co-administration with other vaccines<br />

Imojev can be given at the same time as the yellow fever vaccine, 24 using<br />

separate syringes and separate injection sites.<br />

Co-administration with other vaccines (including other live vaccines) has not<br />

been assessed. If Imojev and the yellow fever vaccine or other live vaccines are<br />

not given simultaneously, they should be given at least 4 weeks apart.<br />

JEspect can be co-administered with the hepatitis A vaccine. 25 Co-administration<br />

with other flavivirus vaccines (including yellow fever vaccine) has not been<br />

assessed.<br />

If co-administration of either JE vaccine with other vaccines is indicated,<br />

injections should be given in separate limbs.<br />

4.8.7 Recommendations<br />

<strong>The</strong> two available JE vaccines are registered for different age groups, and have<br />

different vaccination schedules, booster dose requirements, and contraindications<br />

for use. <strong>The</strong>se factors should be taken into account when deciding the most<br />

appropriate vaccine to use.<br />

<strong>The</strong> dose of Imojev should be administered at least 14 days prior to potential JE<br />

virus exposure.<br />

<strong>The</strong> JEspect vaccine (2-dose) schedule should be completed at least 1 week prior<br />

to potential JE virus exposure.<br />

Travellers<br />

JE vaccination is recommended for:<br />

• travellers (≥12 months of age) spending 1 month or more in rural areas of<br />

high-risk countries in Asia and Papua New Guinea (see 4.8.3 Epidemiology<br />

above); however, as JE has occurred in travellers after shorter periods of<br />

travel, JE vaccination should be considered for shorter-term travellers,<br />

particularly if the travel is during the wet season, or anticipated to be<br />

repeated, and/or there is considerable outdoor activity, and/or the<br />

accommodation is not mosquito-proof26 • all other travellers spending a year or more in Asia (except Singapore), even<br />

if much of the stay is in urban areas.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 263<br />

4.8 JAPANESE ENCEPHALITIS


All travellers to Asia (and other tropical regions) must be fully aware of the need<br />

to take appropriate measures to avoid mosquito bites.<br />

<strong>The</strong> risk of JE to travellers to Asia is determined by the season of travel, the<br />

regions visited, the duration of travel, the extent of outdoor activity and the<br />

extent to which mosquito avoidance measures are taken. 1 Clearly the risk is<br />

greater during prolonged travel to rural areas of Asia during the wet season; it is<br />

probably negligible during short business trips to urban areas.<br />

Torres Strait Islands<br />

JE vaccination is recommended for:<br />

• all residents (≥12 months of age) of the outer islands in the Torres Strait<br />

• all non-residents (≥12 months of age) who will be living or working on the<br />

outer islands of the Torres Strait for a cumulative total of 30 days or more<br />

during the wet season (December to May).<br />

Note: <strong>The</strong> period of greatest risk is from February to March and the vaccination<br />

course should be completed before February. Those arriving in the outer islands<br />

late in the wet season (i.e. in May) will arrive after the risk period and do not<br />

require vaccination. Those visiting the outer islands in the dry season (June to<br />

November) do not require vaccination. Those visiting only the inner islands,<br />

including Thursday Island, do not require vaccination.<br />

Laboratory personnel<br />

JE vaccination is recommended for all research laboratory personnel who will<br />

potentially be exposed to the virus.<br />

Booster doses<br />

Currently there is limited evidence available to inform recommendations<br />

regarding the need and appropriate time interval for a booster dose of either<br />

Imojev or JEspect.<br />

A booster dose of Imojev is not currently recommended. Preliminary data from<br />

a subset of participants in a phase II randomised controlled trial demonstrated<br />

considerable persistence of seroprotective antibody levels 60 months following a<br />

single dose of Imojev12 (see 4.8.4 Vaccines above).<br />

Some data suggest that if the primary series of JEspect was administered >1 year<br />

previously, and there is an ongoing or high risk of JE virus exposure, a booster<br />

dose should be offered. 16<br />

For individuals previously vaccinated with the mouse brain-derived JE vaccine,<br />

either Imojev or JEspect can be used for revaccination if there is an ongoing risk<br />

of JE virus exposure.<br />

264 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.8.8 Pregnancy and breastfeeding<br />

Imojev is a live attenuated viral vaccine and is contraindicated in pregnant<br />

women. Pregnancy should be avoided for 28 days after vaccination.<br />

<strong>The</strong>re are no data on whether the Imojev vaccine virus is excreted in breast milk;<br />

the vaccine should not be given to breastfeeding women.<br />

JEspect vaccine is not routinely recommended for pregnant or breastfeeding<br />

women. However, as JE virus infection during the first and second trimesters<br />

has been associated with miscarriage, pregnant women at risk of acquiring JE<br />

should be offered JE vaccination. Although this inactivated JE vaccine might pose<br />

a theoretical risk to the developing fetus, no adverse outcomes of pregnancy have<br />

been attributed to vaccination against JE.<br />

No specific data are available regarding the administration of JEspect to<br />

breastfeeding women. Breastfeeding women who are at increased risk of<br />

acquiring JE should be offered JE vaccination.<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.8.9 Contraindications<br />

JE vaccines are contraindicated in persons who have had:<br />

• anaphylaxis following a previous dose of any JE vaccine<br />

• anaphylaxis following any vaccine component.<br />

Imojev is a live attenuated viral vaccine and must not be administered to<br />

pregnant or breastfeeding women or to any person who is immunocompromised<br />

due to either disease and/or medical treatment.<br />

4.8.10 Precautions<br />

JE vaccines should not be administered during an acute febrile illness.<br />

<strong>The</strong>re are few data on the safety and efficacy of JEspect vaccine in persons who<br />

are immunocompromised. Such persons may not mount an adequate immune<br />

response, but, as JEspect is an inactivated vaccine, safety and reactogenicity are<br />

not expected to be of concern in those who are immunocompromised.<br />

4.8.11 Adverse events<br />

Local reactions and minor systemic reactions are common to very common<br />

following vaccination against JE.<br />

In adults, adverse events following Imojev were similar to those in placebo<br />

recipients, 9,12 but occurred less often than in recipients of the mouse brainderived<br />

JE vaccine. 9 <strong>The</strong> most common adverse events in two key studies were<br />

injection site pain, headache, fatigue and malaise; most symptoms resolved<br />

within 3 days. 9 Similarly, in children aged 12–24 months, the frequency of<br />

adverse events after Imojev was comparable to that after the hepatitis A vaccine.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 265<br />

4.8 JAPANESE ENCEPHALITIS


About 40% of these subjects reported injection site reactions, including pain<br />

(32%), erythema (23%) and swelling (9%), and about 50% reported at least<br />

one systemic reaction, including fever (21%), appetite loss (26%), irritability<br />

(28%) and abnormal crying (23%). Frequencies of adverse events in children<br />

aged 2–5 years who received Imojev after having been previously vaccinated<br />

with the mouse brain-derived vaccine were similar to, or lower than, those<br />

seen in children aged 12–24 months not previously vaccinated. All reactions<br />

were transient and almost all were mild. Most systemic reactions were mild or<br />

moderate, appeared within 7 days of vaccination, and lasted up to 3 days. 10<br />

In a pooled analysis of over 4000 healthy adults who received at least 1 dose<br />

of JEspect, 54% reported injection site reactions, most commonly pain (33%),<br />

tenderness (33%) and redness (9%). 27 Headache and myalgia were the most<br />

commonly reported systemic adverse events. 17,19,27-29 An earlier analysis found a<br />

comparable rate of adverse events in those who received JEspect compared with<br />

aluminium-containing placebo. 28 Post-marketing surveillance reported adverse<br />

events following JEspect at a rate of about 10 per 100 000 doses distributed;<br />

no serious allergic reactions were observed during the first 12 months after<br />

marketing approval. 27 <strong>The</strong> frequencies of adverse events reported following a<br />

booster dose were similar to those reported after a primary course. 17,18<br />

4.8.12 Public health management of Japanese encephalitis<br />

JE virus infection is a notifiable disease in all states and territories in Australia.<br />

Further instructions about the public health management of JE, including<br />

management of cases of JE, should be obtained from state/territory public<br />

health authorities (see Appendix 1 Contact details for <strong>Australian</strong>, state and territory<br />

government health authorities and communicable disease control)<br />

4.8.13 Variations from product information<br />

JEspect is registered for use in persons aged ≥18 years. <strong>The</strong> ATAGI recommends<br />

that JEspect can be administered to persons aged ≥12 months in circumstances<br />

where an alternative is not available or contraindicated. <strong>The</strong> ATAGI also<br />

recommends that children aged ≥12 months to


4.9 MEASLES<br />

4.9.1 Virology<br />

Measles is a paramyxovirus, genus Morbillivirus. It is an RNA virus with six<br />

structural proteins, three complexed to the RNA and three associated with the<br />

viral envelope. Two of the envelope proteins, the F (fusion) protein and the H<br />

(haemagglutinin) protein, are the most important in pathogenesis. <strong>The</strong> measles<br />

virus can survive for up to 2 hours in air, but is rapidly inactivated by heat, light<br />

and extremes of pH. 1,2<br />

4.9.2 Clinical features<br />

Measles is a highly infectious, acute viral illness spread by respiratory<br />

secretions, including aerosol transmission. 1 It is infectious from the beginning<br />

of the prodromal period and for up to 4 days after the appearance of the rash.<br />

<strong>The</strong> incubation period is usually 10 to 14 days. <strong>The</strong> prodrome, lasting 2 to 4<br />

days, is characterised by fever and malaise, followed by a cough, coryza and<br />

conjunctivitis. <strong>The</strong> maculopapular rash typically begins on the face and upper<br />

neck, and then becomes generalised.<br />

Measles is often a severe disease, frequently complicated by otitis media (in<br />

approximately 9%), pneumonia (in approximately 6%) and diarrhoea (in<br />

approximately 8%). 1,2 Acute encephalitis occurs in 1 per 1000 cases, and has<br />

a mortality rate of 10 to 15%, with a high proportion of survivors suffering<br />

permanent brain damage. 3 Subacute sclerosing panencephalitis (SSPE) is a<br />

late complication of measles, occurring on average 7 years after infection in<br />

approximately 0.5 to 1 per 100 000 measles cases. 1 SSPE causes progressive brain<br />

damage and is always fatal. Complications from measles are more common and<br />

more severe in the chronically ill, in children 95%<br />

PART 4 VACCINE-PREVENTABLE DISEASES 267<br />

4.9 MEASLES


for a single dose and >90% for 2 doses required. 9 A decline in vaccination rates<br />

has resulted in a resurgence in endemic transmission in a number of European<br />

countries, including the United Kingdom. 10 In 2009, the <strong>Australian</strong> Childhood<br />

<strong>Immunisation</strong> Register recorded that 94.0% of children aged 2 years had received<br />

at least 1 dose of measles-containing vaccine and 80.3% of children aged 5 years<br />

had received 2 doses. 11<br />

National serosurveys in early 1999 (evaluating the 1998 National Measles Control<br />

Campaign) and in 2000 showed that those most at risk of measles infection<br />

in Australia were infants


4.9.4 Vaccines<br />

Monovalent measles vaccine is not available in Australia. Measles vaccination<br />

is provided using either measles-mumps-rubella (MMR) or measles-mumpsrubella-varicella<br />

(MMRV) vaccines. Two quadrivalent combination vaccines<br />

containing live attenuated measles, mumps, rubella and varicella viruses<br />

(MMRV) are registered in Australia.<br />

Measles immunity induced by 1-dose vaccination provides long-term immunity<br />

in most recipients. 2,24 However, approximately 5% of recipients fail to develop<br />

immunity to measles after 1 dose. 25 Following a 2nd vaccine dose, approximately<br />

99% of subjects overall will be immune to measles. Measles vaccine effectiveness<br />

studies have found the measles-containing vaccines to be 90 to 95% effective in<br />

developed country settings with high vaccination coverage and low incidence of<br />

measles. 2 A Cochrane review reported 1-dose vaccine effectiveness to be 95%; 26<br />

however, effectiveness has been demonstrated to be lower, particularly by region<br />

(e.g. Asia, Africa) in 1-dose recipients. 27<br />

Combination MMRV vaccines have been shown in clinical trials, predominantly<br />

conducted in children 12 months to 6 years of age, to produce similar rates<br />

of seroconversion to all four vaccine components compared with MMR and<br />

monovalent varicella vaccines administered concomitantly at separate injection<br />

sites. 28-31 In one comparative study assessing seroresponses to a single MMRV<br />

vaccine dose in 12–14-month-old children, the seroresponse rates to measles,<br />

mumps and rubella were similar, but varicella seroresponses were lower in<br />

Priorix-tetra recipients than in ProQuad recipients. 32 However, the clinical<br />

significance of this is not clear, particularly for MMRV given after MMR vaccine. 32<br />

Information on adverse events related to MMR and MMRV vaccines is provided<br />

in 4.9.11 Adverse events below, and also in 4.22 Varicella (for MMRV).<br />

Trivalent measles-mumps-rubella (MMR) vaccines<br />

• M-M-R II – CSL Limited/Merck & Co Inc (live attenuated measles<br />

virus [Enders’ attenuated Edmonston strain], mumps virus [Jeryl<br />

Lynn B level strain] and rubella virus [Wistar RA 27/3 strain]).<br />

Lyophilised pellet in a monodose vial with separate diluent. Each<br />

0.5 mL reconstituted dose contains ≥1000 tissue culture infectious<br />

dose 50% (TCID ) of Enders’ attenuated Edmonston measles<br />

50<br />

virus, ≥12 500 TCID of the Jeryl Lynn B level mumps virus, and<br />

50<br />

≥1000 TCID of the Wistar RA 27/3 rubella virus; sorbitol; sucrose;<br />

50<br />

hydrolysed gelatin; human albumin; fetal bovine serum; neomycin.<br />

• Priorix – GlaxoSmithKline (live attenuated measles virus [Schwarz<br />

strain], mumps virus [RIT 4385 strain, derived from the Jeryl Lynn<br />

strain] and rubella virus [Wistar RA 27/3 strain]). Lyophilised pellet<br />

PART 4 VACCINE-PREVENTABLE DISEASES 269<br />

4.9 MEASLES


in a monodose vial with a pre-filled diluent syringe. Each 0.5 mL<br />

reconstituted dose contains ≥10 3.0 cell culture infectious dose 50%<br />

(CCID 50 ) of the Schwarz measles virus, ≥10 3.7 CCID 50 of the RIT 4385<br />

mumps virus, and ≥10 3.0 CCID 50 of the Wistar RA 27/3 rubella virus;<br />

lactose; neomycin; sorbitol; mannitol.<br />

Quadrivalent measles-mumps-rubella-varicella (MMRV) vaccines<br />

• Priorix-tetra – GlaxoSmithKline (live attenuated measles virus<br />

[Schwarz strain], mumps virus [RIT 4385 strain, derived from the<br />

Jeryl Lynn strain], rubella virus [Wistar RA 27/3 strain] and varicellazoster<br />

virus [Oka strain]). Lyophilised pellet in a monodose vial with<br />

a pre-filled diluent syringe. Each 0.5 mL reconstituted dose contains<br />

≥103.0 CCID of the Schwarz measles virus, ≥10 50 4.4 CCID of the RIT<br />

50<br />

4385 mumps virus, ≥103.0 CCID of the Wistar RA 27/3 rubella virus,<br />

50<br />

and ≥103.3 plaque-forming units (PFU) of Oka varicella-zoster virus;<br />

lactose; neomycin; sorbitol; mannitol.<br />

• ProQuad – CSL Limited/Merck & Co Inc (live attenuated measles<br />

virus [Enders’ attenuated Edmonston strain], mumps virus [Jeryl<br />

Lynn B level strain], rubella virus [Wistar RA 27/3 strain] and<br />

varicella-zoster virus [Oka/Merck strain]). Lyophilised powder<br />

in a monodose vial with a pre-filled diluent syringe. Each 0.5 mL<br />

reconstituted dose contains ≥10 3.0 TCID 50 of Enders’ attenuated<br />

Edmonston measles virus, ≥10 4.3 TCID 50 of the Jeryl Lynn B level<br />

mumps virus, ≥10 3.0 TCID 50 of the Wistar RA 27/3 rubella virus, and<br />

≥10 3.99 PFU of Oka/Merck varicella virus; sucrose; hydrolysed gelatin;<br />

urea; sorbitol; monosodium L-glutamate; human albumin; neomycin;<br />

residual components of MRC-5 cells; bovine serum albumin.<br />

4.9.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 33 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

Both MMR and MMRV vaccines must be reconstituted by adding the entire<br />

contents of the diluent container to the vial containing the pellet and shaking<br />

until the pellet is completely dissolved.<br />

Reconstituted Priorix (MMR), M-M-R II (MMR) and Priorix-tetra (MMRV)<br />

vaccines should be used as soon as practicable. If storage is necessary, hold at<br />

+2°C to +8°C for not more than 8 hours.<br />

Reconstituted ProQuad (MMRV) vaccine must be used within 30 minutes.<br />

270 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.9.6 Dosage and administration<br />

<strong>The</strong> dose of Priorix (MMR) vaccine for both children and adults is 0.5 mL, to be<br />

given by either SC or IM injection.<br />

<strong>The</strong> dose of M-M-R II (MMR) vaccine for both children and adults is 0.5 mL, to<br />

be given by SC injection.<br />

For children


If MMR vaccine is given at


Table 4.9.1: Recommendations for measles vaccination with (a) measlesmumps-rubella<br />

(MMR) (currently available), and (b) once measlesmumps-rubella-varicella<br />

(MMRV) vaccines are available from<br />

July <strong>2013</strong><br />

Vaccines<br />

(a) Only monovalent varicella<br />

vaccine available<br />

(b) When MMRV vaccine available<br />

(from July <strong>2013</strong>)<br />

Schedule point (age)<br />

12 months 18 months 4 years<br />

MMR VV MMR *<br />

MMR MMRV –<br />

* <strong>The</strong> 2nd dose of MMR-containing vaccine is recommended to be provided at 18 months of age<br />

to improve 2-dose coverage and protection against measles in young children. However, until<br />

June <strong>2013</strong>, the 2nd dose of MMR vaccine is included under the NIP schedule for administration<br />

at 4 years of age. From July <strong>2013</strong>, the 2nd dose of MMR vaccine will move to the 18-month NIP<br />

schedule point and be provided as MMRV vaccine.<br />

Adults and adolescents<br />

Persons born before 1966<br />

No vaccination is required for persons born before 1966 (unless serological<br />

evidence indicates otherwise), as circulating virus and disease were prevalent<br />

before this time, suggesting most persons would have acquired immunity<br />

from natural infection. However, confirmed cases of measles have occurred in<br />

persons born before 19667 and, if doubt exists, it may be more expedient to offer<br />

vaccination than serological testing. (See also ‘Serological testing for immunity to<br />

measles’ below.)<br />

Persons born during or since 1966<br />

All persons born during or since 1966 who are ≥18 months of age (or, until<br />

catch-up following the move of the 2nd NIP dose of measles-containing<br />

vaccine to 18 months of age is completed, are ≥4 years of age) should have<br />

documented evidence of 2 doses of MMR-containing vaccine (administered<br />

at least 4 weeks apart and with both doses administered at ≥12 months of age;<br />

see ‘Children’ above) or have serological evidence of protection for measles,<br />

mumps and rubella.<br />

It is recommended that all adolescents and young adults have their<br />

vaccination records reviewed to ensure they have received 2 doses of MMR<br />

vaccine (see 4.9.3 Epidemiology above).<br />

MMRV vaccines are not recommended for use in persons ≥14 years of age, due<br />

to a lack of data on safety and immunogenicity/efficacy in this age group. If a<br />

dose of MMRV vaccine is inadvertently given to an older person, this dose does<br />

not need to be repeated.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 273<br />

4.9 MEASLES


Healthcare workers and other occupations<br />

All adolescents and adults (born during or since 1966) should have their<br />

vaccination records reviewed to ensure they have received 2 doses of MMR<br />

vaccine. This is important for all persons, but especially those working in certain<br />

occupations, such as healthcare workers, staff working in early childhood<br />

education and care, staff of long-term care facilities and staff of correctional<br />

facilities. Those who were born during or since 1966 and are non-immune, or<br />

who have only received 1 dose of MMR vaccine, should be vaccinated and have<br />

documented evidence of 2 doses of MMR vaccine or serological evidence of<br />

immunity to measles (see ‘Adults and adolescents’ above). (See also 3.3 Groups<br />

with special vaccination requirements, Table 3.3.7 Recommended vaccinations for<br />

persons at increased risk of certain occupationally acquired vaccine-preventable diseases.)<br />

Travellers<br />

It is especially important that all persons born during or since 1966 have been<br />

given 2 doses of measles-containing vaccine (administered at least 4 weeks apart,<br />

with both doses administered at ≥12 months of age; see ‘Children’ above) before<br />

embarking on international travel if they do not have evidence of previous<br />

receipt of 2 doses of MMR vaccine or serological evidence of protection for<br />

measles, mumps and rubella (see ‘Adults and adolescents’ above).<br />

Infants travelling to countries in which measles is endemic, or where measles<br />

outbreaks are occurring, may be given MMR vaccine from as young as 9 months<br />

of age, after an individual risk assessment. In these cases, 2 further doses of<br />

MMR vaccine are still required. <strong>The</strong> next dose of MMR vaccine should be given<br />

at 12 months of age or 4 weeks after the 1st dose, whichever is later. This should<br />

be followed by the routine administration of the next dose of measles-containing<br />

vaccine, as MMRV vaccine, at 18 months of age.<br />

Serological testing for immunity to measles<br />

Serological testing for immunity to measles, mumps, rubella or varicella is not<br />

recommended before or after routine administration of the 2-dose childhood<br />

schedule of these vaccines.<br />

However, serological testing for measles can be performed in cases where a<br />

history of natural immunity or 2 doses of vaccine administration is uncertain<br />

(see ‘Adults and adolescents’ above). Serology is indicated in special situations,<br />

such as pre-pregnancy planning (see also 4.18 Rubella, 4.22 Varicella and 3.3.2<br />

Vaccination of women who are planning pregnancy, pregnant or breastfeeding, and<br />

preterm infants). Serological tests to investigate immunity to measles are generally<br />

sensitive at detecting antibody produced by both prior natural infection and<br />

vaccination, although sensitivity varies by assay and the clinical setting (e.g.<br />

time since vaccination). 2 Interpretation of the results of serological testing may<br />

be enhanced by discussion with the laboratory that performed the test, ensuring<br />

that relevant clinical information is provided. An alternative to serological testing<br />

is presumptive administration of MMR vaccine dose(s). <strong>The</strong>re is no known<br />

274 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


increase in adverse events from vaccinating those with pre-existing immunity to<br />

one or more of the vaccine components (see 4.9.11 Adverse events below).<br />

4.9.8 Pregnancy and breastfeeding<br />

MMR-containing vaccines are contraindicated in pregnant women. Pregnancy<br />

should be avoided for 28 days after vaccination. 38<br />

MMR vaccines can be given to breastfeeding women. (See also 4.18 Rubella.)<br />

MMRV vaccines are not recommended for use in persons aged ≥14 years.<br />

<strong>The</strong>re is no risk to pregnant women from contact with recently vaccinated<br />

persons. <strong>The</strong> vaccine virus is not transmitted from vaccinated persons to<br />

susceptible contacts. 1<br />

See also 4.18 Rubella, 4.22 Varicella and 3.3 Groups with special vaccination<br />

requirements, Table 3.3.1 Recommendations for vaccination in pregnancy for more<br />

information.<br />

4.9.9 Contraindications<br />

Anaphylaxis to vaccine components<br />

MMR and MMRV vaccines are contraindicated in persons who have had:<br />

• anaphylaxis following a previous dose of any MMR-containing vaccine<br />

• anaphylaxis following any vaccine component.<br />

Persons who are immunocompromised<br />

Measles-, mumps- and rubella-containing vaccines contain live<br />

attenuated vaccine viruses and are contraindicated in persons who are<br />

immunocompromised. Thus, MMR-containing vaccines are contraindicated in<br />

the following groups:<br />

• Persons immunocompromised due to HIV/AIDS. MMR vaccination of<br />

asymptomatic HIV-infected persons >12 months of age with an age-specific<br />

CD4 + count of ≥15% may be considered39-42 (see ‘HIV-infected persons’<br />

in 3.3.3 Vaccination of immunocompromised persons). Since studies have not<br />

been performed using combination MMRV vaccines in asymptomatic HIVinfected<br />

persons or persons with an age-specific CD4 + count of ≥15%, it is<br />

recommended that only MMR vaccine (and monovalent VV, see 4.22 Varicella)<br />

be considered for use in this setting. 41,43-45<br />

• Persons with other medical conditions associated with significant<br />

immunocompromise (see 3.3.3 Vaccination of immunocompromised persons).<br />

• Persons receiving high-dose systemic immunosuppressive therapy,<br />

such as chemotherapy, radiation therapy or oral corticosteroids. MMRcontaining<br />

vaccines are contraindicated in persons taking high-dose oral<br />

corticosteroids for more than 1 week (in children equivalent to >2 mg/kg<br />

per day prednisolone, and in adults >60 mg per day) (see 3.3.3 Vaccination<br />

PART 4 VACCINE-PREVENTABLE DISEASES 275<br />

4.9 MEASLES


of immunocompromised persons). Those who have been receiving highdose<br />

systemic steroids for more than 1 week may be vaccinated with live<br />

attenuated vaccines after corticosteroid therapy has been discontinued<br />

for at least 1 month46 (see 4.9.10 Precautions below and 3.3.3 Vaccination of<br />

immunocompromised persons).<br />

See also 3.3 Groups with special vaccination requirements and 4.22 Varicella for more<br />

information.<br />

Pregnant women<br />

See 4.9.8 Pregnancy and breastfeeding above.<br />

4.9.10 Precautions<br />

Persons with egg allergy<br />

Children with egg allergy can be safely given MMR or MMRV vaccine. 1,47 Skin<br />

testing is not required prior to vaccine administration. 1 Although measles and<br />

mumps (but not rubella or varicella) vaccine viruses are grown in chick embryo<br />

tissue cultures, it is now recognised that measles- and mumps-containing<br />

vaccines contain negligible amounts of egg ovalbumin (see 4.9.13 Variations from<br />

product information below).<br />

Vaccination with other live attenuated parenteral vaccines<br />

If MMR or MMRV vaccine is not given simultaneously with other live attenuated<br />

parenteral vaccines (e.g. varicella, BCG, yellow fever), the vaccines should be<br />

given at least 4 weeks apart.<br />

Vaccination after immunoglobulin or blood product administration<br />

Administration of MMR or MMRV vaccine should be delayed after<br />

administration of immunoglobulin-containing products. After receipt of<br />

immunoglobulin-containing blood products, the expected immune response<br />

to measles, mumps, rubella and varicella vaccination may be impaired. 25,35,48<br />

MMR-containing vaccines should not be given for between 3 and 11 months<br />

following the administration of immunoglobulin-containing blood products.<br />

<strong>The</strong> interval between receipt of the blood product and vaccination depends on<br />

the amount of immunoglobulin in each product, and is indicated in 3.3 Groups<br />

with special vaccination requirements, Table 3.3.6 Recommended intervals between<br />

either immunoglobulins or blood products and MMR, MMRV or varicella vaccination. 35<br />

For further information, see 3.3.4 Vaccination of recent recipients of normal human<br />

immunoglobulin and other blood products.<br />

Recent blood transfusion with washed red blood cells is not a contraindication to<br />

MMR or MMRV vaccines.<br />

MMR vaccine may be administered concomitantly with, or at any time in<br />

relation to, anti-D immunoglobulin, but at a separate injection site. Anti-D<br />

immunoglobulin does not interfere with the antibody response to vaccine.<br />

276 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Immunoglobulin or blood product administration after vaccination<br />

Immunoglobulin-containing products should not be administered for 3 weeks<br />

following vaccination with measles-containing vaccines, unless the benefits<br />

exceed those of vaccination. If immunoglobulin-containing products are<br />

administered within this interval, the vaccinated person should either be<br />

revaccinated later at the appropriate time following the product (as indicated in<br />

Table 3.3.6), or be tested for immunity 6 months later and then revaccinated if<br />

seronegative.<br />

Rh (D) immunoglobulin (anti-D) may be given at the same time in different sites<br />

with separate syringes or at any time in relation to MMR vaccine, as it does not<br />

interfere with the antibody response to the vaccine.<br />

HIV-infected persons<br />

MMR vaccine can be given to asymptomatic HIV-infected persons >12 months<br />

of age with an age-specific CD4 + count of ≥15% 49 (see 3.3 Groups with special<br />

vaccination requirements, Table 3.3.4 Categories of immunocompromise in HIV-infected<br />

persons, based on age-specific CD4 + counts and percentage of total lymphocytes). This is<br />

because the risk posed by measles infection is considered to be greater than the<br />

likelihood of adverse events from vaccination. 46 MMR vaccine is contraindicated<br />

in immunocompromised HIV-infected persons (see 4.9.9 Contraindications above).<br />

As there are no data available on the safety, immunogenicity or efficacy of MMRV<br />

vaccines in HIV-infected children, MMRV vaccine should not be administered as<br />

a substitute for MMR vaccine when vaccinating these children. 25,48<br />

Persons receiving immunosuppressive therapy<br />

MMR-containing vaccines may be given to persons on low-dose systemic<br />

corticosteroid therapy (e.g. children on doses of ≤2 mg/kg per day for less than<br />

1 week, and those on lower doses of 1 mg/kg per day or alternate-day regimens<br />

for longer periods). Persons receiving high-dose corticosteroids can receive<br />

MMR-containing vaccines after corticosteroid therapy has been discontinued<br />

for at least 1 month (see 4.9.9 Contraindications above). 46 Some experts<br />

suggest withholding lower doses of steroids 2 to 3 weeks prior to vaccination<br />

with live viral vaccines, if this is possible. 46,48 (See also 3.3.3 Vaccination of<br />

immunocompromised persons.)<br />

Household contacts of persons who are immunocompromised<br />

Household contacts of persons who are immunocompromised, should ensure<br />

that they are age-appropriately vaccinated against, or are immune to, measles,<br />

as well as mumps, rubella and varicella. MMR-containing vaccines can be safely<br />

administered to household contacts, as measles, mumps and rubella vaccine<br />

viruses are not transmissible from vaccinated persons to others. 25 If using MMRV<br />

vaccine, see 4.22 Varicella for information regarding varicella vaccine virus<br />

transmission.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 277<br />

4.9 MEASLES


Persons receiving long-term aspirin or salicylate therapy<br />

<strong>The</strong>re is no need to avoid salicylates before or after MMR or MMRV vaccination.<br />

Persons receiving long-term salicylate therapy (aspirin) can be vaccinated with<br />

MMRV, if indicated, as the benefit is likely to outweigh any possible risk of Reye<br />

syndrome occurring after vaccination with a varicella-containing vaccine (see<br />

4.22 Varicella).<br />

Persons with a history of thrombocytopenia<br />

Thrombocytopenia is a rare adverse event following MMR vaccination (see<br />

also 4.9.11 Adverse events below). 1,50,51 In children with a past history of an<br />

episode(s) of idiopathic thrombocytopenia purpura (ITP), the risk of vaccineassociated<br />

thrombocytopenia occurring following a dose of MMR vaccine has<br />

been uncertain. 25,51 However, a recent systematic review concluded that MMR<br />

vaccination, either as a 1st or 2nd dose, did not lead to a recurrence of ITP. 52<br />

Personal or close family history of seizures or convulsions<br />

Children with a personal or close family history of seizures or convulsions<br />

should be given MMR or MMRV vaccine, provided the parents/carers<br />

understand that there may be a febrile response 5 to 12 days after vaccination. 25<br />

Advice should be given regarding reducing fever with paracetamol and other<br />

measures. Due to an increased risk of fever and febrile convulsions in 1st dose<br />

recipients of MMRV vaccine, MMRV vaccines are only recommended for use as<br />

the 2nd dose of MMR-containing vaccine (see 4.9.7 Recommendations above and<br />

4.9.11 Adverse events below).<br />

Tuberculin skin testing following MMR vaccination<br />

Measles virus inhibits the response to tuberculin and tuberculin-positive persons<br />

may become tuberculin-negative for up to a month after measles infection. 25,53 As<br />

such, tuberculin skin testing (Mantoux test) may be unreliable for at least 4 weeks<br />

after the administration of measles-containing vaccines. <strong>The</strong>re are no studies<br />

on the effect of MMR or MMRV vaccination on the results of interferon-gamma<br />

release assays (IGRAs). 54<br />

4.9.11 Adverse events<br />

If using MMRV vaccine, additional adverse events relating to the varicella<br />

vaccine component are outlined in 4.22 Varicella.<br />

Adverse events following administration of MMR-containing vaccines are<br />

generally mild and well tolerated. 2 Adverse events are much less common after<br />

the 2nd dose of MMR or MMRV vaccine than after the 1st dose.<br />

Fever (with malaise and/or a rash, which is non-infectious) may occur after<br />

MMR vaccination, most commonly between 7 to 10 days (range 5 to 12<br />

days) after vaccination and lasting about 2 to 3 days. This coincides with the<br />

period of peak measles vaccine virus replication. High fever (>39.4°C) occurs<br />

in approximately 5 to 15% of MMR vaccine recipients, and rash occurs in<br />

278 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


approximately 5%. 2,25 <strong>The</strong>re is also an increased risk for febrile seizures in the<br />

same time period of approximately 1 case per 3000 doses. 25<br />

It is recommended that vaccine recipients or their parents/carers be advised<br />

about possible symptoms, and given advice for reducing fever, including the use<br />

of paracetamol for fever in the period 5 to 12 days after vaccination.<br />

Higher rates of fever were observed in clinical trials of both MMRV vaccines,<br />

particularly following dose 1, when compared with giving MMR vaccine and<br />

monovalent VV at the same time but at separate sites. 28-31 Two post-marketing<br />

studies in the United States identified an approximately 2-fold increased risk of<br />

fever and febrile convulsions in 1st dose recipients of MMRV vaccine, who were<br />

predominantly 12–23 months of age, in the period 7 to 10 days55 (or 5 to 12 days) 56<br />

after vaccination, compared with recipients of separate MMR and VV vaccines.<br />

MMRV vaccination resulted in 1 additional febrile seizure for every 2300 doses<br />

compared to separate MMR and VV vaccination. 55 An increase in fever or febrile<br />

convulsions has not been identified after the 2nd dose of MMRV vaccine in the<br />

United States, although most 2nd dose recipients were aged 4–6 years, an age at<br />

which the incidence of febrile convulsions is low. 57 <strong>The</strong>se post-marketing studies<br />

were in children who received ProQuad; however, it is anticipated that this side<br />

effect profile would be similar in Priorix-tetra recipients.<br />

A varicelliform rash may occur after MMRV vaccination (see 4.22.11 Adverse<br />

events in 4.22 Varicella). <strong>The</strong> appearance of a rash after monovalent varicella<br />

vaccine occurs in less than 5% of vaccine recipients (usually within 5 to 26 days),<br />

and similar rates are observed with the use of MMRV vaccine. 58<br />

Anaphylaxis following the administration of MMR vaccine is very rare (less<br />

than 1 in 1 million doses distributed). 25 Although no cases of anaphylaxis were<br />

reported in MMRV vaccine clinical trials, the incidence is likely to be similar to<br />

that occurring with use of MMR vaccine. Anaphylaxis after vaccination is likely<br />

due to anaphylactic sensitivity to gelatin or neomycin, not egg allergy (see 4.9.10<br />

Precautions above).<br />

Thrombocytopenia (usually self-limiting) has been very rarely associated with the<br />

rubella or measles component of MMR vaccine, occurring in 3 to 5 per 100 000<br />

doses of MMR vaccine administered. 1,25,50,51 This is considerably less frequent<br />

than after natural measles, mumps and rubella infections. 51 Any association with<br />

MMRV vaccine is expected to be similar.<br />

It is uncertain whether encephalopathy occurs after measles vaccination. If it<br />

does, it is at least 1000 times less frequent than as a complication from natural<br />

infection. 2,25<br />

Other rare adverse events attributed to MMR vaccine include transient<br />

lymphadenopathy and arthralgia (see 4.18 Rubella). Parotitis has been reported<br />

rarely (see 4.11 Mumps). 25<br />

PART 4 VACCINE-PREVENTABLE DISEASES 279<br />

4.9 MEASLES


Autism, autistic spectrum disorder and inflammatory bowel disease are<br />

not associated with the MMR vaccine. <strong>The</strong>re has been no credible scientific<br />

evidence to support this claim and most proponents of the link have retracted<br />

this claim. 59,60 <strong>The</strong>re is now a substantial body of evidence to refute it61-64 (see<br />

Appendix 4 Commonly asked questions about vaccination).<br />

4.9.12 Public health management of measles<br />

Measles is a notifiable disease in all states and territories in Australia. <strong>The</strong> public<br />

health management of measles is described in Measles: national guidelines for public<br />

health units37 (www.health.gov.au/cdnasongs) and is given urgent public health<br />

priority. Refer to the national guidelines for current case definitions, testing and<br />

post-exposure prophylaxis of contacts.<br />

Further instructions about the public health management of measles can also<br />

be obtained from state/territory public health authorities (see Appendix 1<br />

Contact details for <strong>Australian</strong>, state and territory government health authorities and<br />

communicable disease control).<br />

MMR vaccine (and MMRV in some instances) is recommended for post-exposure<br />

prophylaxis within 72 hours of a non-immune individual being exposed to<br />

measles. See Table 4.9.2 for detailed information. Administration of normal<br />

human immunoglobulin (NHIG), rather than MMR or MMRV, is recommended<br />

in some settings (see Part 5 Passive immunisation and Table 4.9.2). 37 Post-exposure<br />

prophylaxis should be given on the direction of public health authorities.<br />

Children >12 months of age who have received 1 dose of measles-containing<br />

vaccine can be offered their 2nd dose early (if at least 4 weeks after the 1st dose<br />

has elapsed) if they are considered at risk of coming in contact with measles37 (see 4.9.7 Recommendations above and Table 4.9.2). If varicella vaccination is also<br />

indicated, MMRV vaccine can be used, although MMRV vaccine is not routinely<br />

recommended as the 1st dose of MMR-containing vaccine in children aged<br />


Table 4.9.2: Post-exposure prophylaxis ≤72 hours since exposed to measles<br />

for non-immune individuals (adapted from Measles: national<br />

guidelines for public health units) 37<br />

Age or immune status Measles-mumps-rubella (MMR) vaccination history<br />

0 doses MMR or<br />

unknown<br />

1 dose MMR 2 doses MMR<br />

Normal human NHIG<br />

NHIG<br />

Immunocompromised<br />

(any age)<br />

immunoglobulin<br />

(NHIG)<br />

0.5 mL/kg to<br />

maximum of<br />

15 mL<br />

0.5 mL/kg to<br />

maximum of<br />

15 mL<br />

0.5 mL/kg to<br />

maximum of<br />

15 mL<br />

NHIG<br />

0.2 mL/kg<br />

only if mother<br />

Not applicable Not applicable<br />

Birth to 5 months<br />

has had


4.9.13 Variations from product information<br />

<strong>The</strong> product information for MMR and MMRV vaccines recommends that<br />

women of child-bearing age should be advised not to become pregnant for<br />

3 months after vaccination. <strong>The</strong> ATAGI instead recommends avoiding<br />

pregnancy for 28 days after vaccination. 38<br />

<strong>The</strong> product information for Priorix, M-M-R II, Priorix-tetra and ProQuad states<br />

that persons with a history of anaphylactic or anaphylactoid reactions to egg<br />

should not be vaccinated. <strong>The</strong> ATAGI recommends instead that either Priorix,<br />

M-M-R II, Priorix-tetra or ProQuad can be given in this situation. 25<br />

<strong>The</strong> product information for Priorix-tetra states that it should be given by SC<br />

injection. <strong>The</strong> ATAGI recommends that it may also be given by IM injection.<br />

<strong>The</strong> product information for ProQuad states that this vaccine is indicated for<br />

vaccination in individuals 12 months through 12 years of age. <strong>The</strong> product<br />

information for Priorix-tetra states that this vaccine can be used in persons from<br />

9 months of age. <strong>The</strong> ATAGI recommends instead that both MMRV vaccines<br />

can be given to persons up to 14 years of age. <strong>The</strong> ATAGI also recommends that<br />

MMRV vaccine should not be used routinely as the 1st dose of MMR-containing<br />

vaccine in children aged


4.10 MENINGOCOCCAL DISEASE<br />

4.10.1 Bacteriology<br />

Meningococcal disease is caused by the bacterium Neisseria meningitidis (or<br />

meningococcus), a Gram-negative diplococcus. <strong>The</strong>re are 13 known serogroups<br />

distinguished by differences in surface polysaccharides of the outer membrane<br />

capsule. Meningococcal serogroups are designated by letters of the alphabet.<br />

Globally, serogroups A, B, C, W 135 and Y most commonly cause disease.<br />

Meningococci can be further classified by differences in their outer membrane<br />

proteins, which are referred to as serotypes and serosubtypes. 1 More recently,<br />

molecular typing has been used to further differentiate meningococci. <strong>The</strong>re is no<br />

consistent relationship between serogroup or serotype/subtype and virulence. 2<br />

4.10.2 Clinical features<br />

N. meningitidis can cause meningitis, septicaemia or a combination of the two.<br />

Other localised infections, including pneumonia, arthritis and conjunctivitis,<br />

may also occur but are uncommon. Septicaemia, with or without meningitis,<br />

can be particularly severe. <strong>The</strong> overall mortality risk for invasive disease is high<br />

(between 5 and 10%), despite appropriate antibiotic therapy. 2 Of those who<br />

survive, approximately 10 to 20% develop permanent sequelae, including limb<br />

deformity, skin scarring and neurologic deficits. 1 Prior invasive meningococcal<br />

disease does not induce protective immunity against the implicated serogroup of<br />

meningococci. <strong>The</strong>refore, persons with a history of meningococcal disease should<br />

still be vaccinated if required.<br />

N. meningitidis is carried and transmitted only by humans. <strong>The</strong>re are no known<br />

animal reservoirs. Asymptomatic respiratory tract carriage of meningococci<br />

is present in about 10% of the population, and the prevalence may be higher<br />

when groups of people occupy small areas of living space. 3,4 Recent studies<br />

indicate that there may be a number of factors that contribute to the increased<br />

risk of contracting meningococcal disease, including exposure to smokers, recent<br />

illness, living in crowded conditions and multiple intimate kissing partners. 2-4<br />

Persons with inherited disorders of phagocytosis associated with properdin<br />

deficiency or absence of the terminal components of complement, as well<br />

as persons with functional or anatomical asplenia, have an increased risk of<br />

meningococcal infection. 1<br />

<strong>The</strong> disease is transmitted via droplets and has an incubation period of between<br />

1 and 10 days, but commonly 3 to 4 days. 4 <strong>The</strong> capacity of meningococcal disease<br />

to have a fulminant and rapidly fatal course in previously healthy (and usually<br />

young) persons causes it to be greatly feared. Intensive public health follow-up<br />

is required after each single case to trace contacts and to institute appropriate<br />

public health measures for them. As a result of all these factors, this disease can<br />

cause widespread community alarm and generate significant media interest. 4<br />

PART 4 VACCINE-PREVENTABLE DISEASES 283<br />

4.10 MENINGOCOCCAL DISEASE


4.10.3 Epidemiology<br />

Meningococci cause both sporadic and epidemic disease throughout the world.<br />

Serogroup A disease occurs predominantly in low-income countries, such as<br />

those in Africa and Asia, while serogroup B is the major cause of sporadic<br />

meningococcal disease in most developed countries, including Australia.<br />

Serogroup C meningococci have been occasionally associated with small<br />

clusters of meningococcal disease cases in schools, universities and nightclubs in<br />

Australia in the past. 5-7 Rarely, there are clusters of meningococcal disease cases<br />

associated with serogroup B. 8<br />

As in other temperate climates, meningococcal disease cases occurring in<br />

Australia tend to follow a seasonal trend, with a large proportion of cases<br />

reported during late winter and early spring. <strong>The</strong> overall notification rate for<br />

invasive meningococcal disease to the National Notifiable Diseases Surveillance<br />

System reached a peak of 3.5 per 100 000 in 2001, but declined to 1 case per<br />

100 000 in 2010. 9,10 <strong>The</strong>re have been considerable differences noted in the<br />

incidence of meningococcal disease between <strong>Australian</strong> states and territories in<br />

the past. Notifications include meningococcal disease cases that were diagnosed<br />

on clinical grounds alone, and those cases that were confirmed by laboratory<br />

methods such as culture, serology or nucleic acid testing of clinical material.<br />

In 2009, 259 cases were reported nationally, of which 194 were laboratoryconfirmed.<br />

9,10 <strong>The</strong> majority of laboratory-confirmed meningococcal cases were<br />

serogroup B (83%) and serogroup C (5.6%). 10 <strong>The</strong>re has been a sustained decline<br />

in serogroup C meningococcal disease among the 1–19 years age group, as<br />

well as other age groups not targeted in the vaccine program, since the 2003<br />

introduction of routine serogroup C vaccination and catch-up programs. 9-11 In<br />

other countries that introduced a meningococcal C vaccination program, this<br />

herd immunity effect has also resulted in a reduction in incidence in age groups<br />

not targeted by the program. 12-14<br />

Meningococcal disease can occur in any age group, but a large proportion of<br />

cases occur in those


4.10.4 Vaccines<br />

<strong>The</strong>re are different types of meningococcal vaccines:<br />

• meningococcal C conjugate vaccines (MenCCV)<br />

• Haemophilus influenzae type b–meningococcal C combination vaccine (Hib-<br />

MenCCV)<br />

• quadrivalent meningococcal conjugate vaccines (4vMenCV)<br />

• quadrivalent meningococcal polysaccharide vaccines (4vMenPV).<br />

Conjugate vaccines<br />

Meningococcal C conjugate vaccines (MenCCV)<br />

• Meningitec – Pfizer Australia Pty Ltd (meningococcal serogroup<br />

C–CRM conjugate). Each 0.5 mL pre-filled syringe contains 10 µg<br />

197<br />

Neisseria meningitidis serogroup C oligosaccharide conjugated to<br />

approximately 15 µg of non-toxic Corynebacterium diphtheriae CRM197 protein; aluminium phosphate.<br />

• Menjugate Syringe – CSL Limited/Novartis Vaccines and Diagnostics<br />

Pty Ltd (meningococcal serogroup C–CRM 197 conjugate). Lyophilised<br />

powder in a monodose vial with a pre-filled diluent syringe. Each<br />

0.5 mL reconstituted dose contains 10 µg N. meningitidis serogroup C<br />

oligosaccharide conjugated to 12.5–25 µg of non-toxic C. diphtheriae<br />

CRM 197 protein; 1.0 mg aluminium hydroxide.<br />

• NeisVac-C – Baxter Healthcare (meningococcal serogroup C–tetanus<br />

toxoid conjugate). Each 0.5 mL pre-filled syringe contains 10 µg<br />

N. meningitidis serogroup C polysaccharide conjugated to 10–20 µg of<br />

tetanus toxoid; 0.5 mg aluminium as aluminium hydroxide.<br />

Combination vaccine that contains meningococcal C<br />

• Menitorix – GlaxoSmithKline (Haemophilus influenzae type b [PRP-T]meningococcal<br />

serogroup C–tetanus toxoid conjugate). Lyophilised<br />

powder in a monodose vial with a pre-filled diluent syringe. Each<br />

0.5 mL reconstituted dose contains 5 µg Hib capsular polysaccharide<br />

(PRP) conjugated to 12.5 µg tetanus toxoid, and 5 µg N. meningitidis<br />

serogroup C polysaccharide conjugated to 5 µg tetanus toxoid; traces<br />

of trometamol and sucrose.<br />

Quadrivalent meningococcal conjugate vaccines (4vMenCV)<br />

• Menactra – Sanofi Pasteur Pty Ltd (meningococcal serogroups A,<br />

C, W , Y–diphtheria toxoid conjugate). Each 0.5 mL monodose vial<br />

135<br />

PART 4 VACCINE-PREVENTABLE DISEASES 285<br />

4.10 MENINGOCOCCAL DISEASE


contains 4 µg each of serogroups A, C, W 135 and Y polysaccharides<br />

conjugated with a total of approximately 48 µg of a diphtheria toxoid<br />

protein.<br />

• Menveo – CSL Limited/Novartis Vaccines and Diagnostics Pty<br />

Ltd (meningococcal serogroups A, C, W 135 , Y–CRM 197 conjugate).<br />

Lyophilised powder containing serogroup A (MenA) in a monodose<br />

vial with a pre-filled syringe or vial containing serogroups C, W 135 and<br />

Y (MenCWY) in saline suspension. Each 0.5 mL reconstituted dose<br />

contains 10 µg of serogroup A and 5 µg each of serogroups C, W 135<br />

and Y oligosaccharides individually conjugated with up to 33.3 µg of<br />

non-toxic C. diphtheriae CRM 197 protein; sucrose.<br />

All conjugate vaccines induce immunity within 10 to 14 days of administration.<br />

MenCCVs confer protection only against serogroup C disease. 4vMenCVs will<br />

provide protection against four serogroups of meningococci: serogroups A, C,<br />

W and Y. Neither MenCCV nor 4vMenCV will provide protection against<br />

135<br />

serogroup B meningococcal disease.<br />

MenCCVs have been used in an infant schedule in many countries, including<br />

Australia. 11-13,17-19 <strong>The</strong> vaccine effectiveness following 1 dose of MenCCV has<br />

been estimated to range from 83 to 100%. 13,17 <strong>The</strong>re has been a sustained decline<br />

in serogroup C meningococcal disease, which has also been observed in age<br />

groups not targeted in vaccination programs, both in Australia and overseas. 11,14<br />

Duration of immunity is still uncertain. However, current serogroup C<br />

meningococcal disease epidemiology in Australia suggests ongoing protection in<br />

those groups previously vaccinated. 11<br />

Hib-MenCCV can be administered where a booster dose of H. influenzae<br />

type b and primary vaccination for meningococcal serogroup C is required.<br />

<strong>The</strong> immunogenicity and safety of Hib-MenCCV as a booster dose has been<br />

demonstrated in clinical trials and in the United Kingdom, where this vaccine is<br />

now administered as part of the infant schedule. 20-25<br />

Several clinical trials have demonstrated the immunogenicity of 4vMenCV,<br />

with human serum bactericidal assay titres of ≥1:4 reported in adolescents and<br />

young adults. 26-28 <strong>The</strong>re have been a number of studies examining 4vMenCV in<br />

children. 29-31 All studies indicated that 4vMenCVs were safe and immunogenic in<br />

both infants and children. 29-33 Menveo has demonstrated superiority to Menactra<br />

in serum bactericidal assays to serogroups A, W and Y (and variably also to<br />

135<br />

serogroup C); however, the clinical relevance of this is currently unknown. 27,34-36<br />

286 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Polysaccharide vaccines<br />

Quadrivalent meningococcal polysaccharide vaccines (4vMenPV)<br />

• Mencevax ACWY – GlaxoSmithKline (meningococcal serogroups A,<br />

C, W 135 and Y polysaccharides). Lyophilised pellet in a monodose vial<br />

with separate saline diluent. Each 0.5 mL reconstituted dose contains<br />

50 µg of each meningococcal serogroup polysaccharide; 12.6 mg<br />

sucrose; 0.1 mg trometamol.<br />

• Menomune – Sanofi Pasteur Pty Ltd (meningococcal serogroups A, C,<br />

W 135 and Y polysaccharides). Lyophilised powder in a monodose vial<br />

with separate saline diluent. Each 0.5 mL reconstituted dose contains<br />

50 µg of each meningococcal serogroup polysaccharide; 2.5–5 mg<br />

lactose.<br />

4vMenPV provides protection against serogroups A, C, W and Y. <strong>The</strong>se<br />

135<br />

vaccines induce antibodies in 10 to 14 days in 90% of recipients >2 years of age.<br />

Immunity decreases markedly during the first 3 years following a single dose of<br />

vaccine, particularly in infants and young children. However, clinical protection<br />

persists for at least 3 years in school-aged children and adults.<br />

<strong>The</strong> duration of immunity is further complicated by the induction of<br />

immunological hyporesponsiveness to the serogroup C component<br />

following repeated vaccination with 4vMenPV, as revaccination results in<br />

a reduced antibody response compared with the primary immunisation. 37<br />

This phenomenon has been noted in both children and adults. 38-40 <strong>The</strong><br />

demonstration of subsequent hyporesponsiveness has led to the concern that<br />

vaccinating persons at low risk may reduce the effectiveness of revaccination<br />

in a subsequent high-risk situation, although this has not been clinically<br />

demonstrated. This hyporesponsiveness can be overcome with meningococcal<br />

conjugate vaccines, 38 although additional doses of a conjugate vaccine may<br />

be required in young children. <strong>The</strong>re is little response to the serogroup C<br />

component of the 4vMenPV before 18 months of age and little response to<br />

serogroup A before 3 months of age. 41<br />

4.10.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 42 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

Conjugate vaccines<br />

Menjugate Syringe must be reconstituted by adding the entire contents of the<br />

diluent syringe to the vial and shaking until the powder is completely dissolved.<br />

Reconstituted vaccine should be used immediately.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 287<br />

4.10 MENINGOCOCCAL DISEASE


<strong>The</strong> product information for NeisVac-C states that this vaccine can be stored at<br />

+25°C for a period of up to 9 months. Refer to product information for further vaccine<br />

storage details.<br />

Menitorix must be reconstituted by adding the entire contents of the diluent<br />

syringe to the vial and shaking until the powder is completely dissolved.<br />

Reconstituted vaccine should be used as soon as practicable. If storage is<br />

necessary, hold at +2°C to +8°C for not more than 24 hours.<br />

Menveo must be reconstituted by adding the entire contents of the liquid<br />

MenCWY syringe/vial to the lyophilised MenA vial and shaking until the<br />

powder is completely dissolved. Reconstituted vaccine should be used as soon<br />

as practicable. If storage is necessary, hold at +2°C to +8°C for not more than<br />

24 hours.<br />

Polysaccharide vaccines<br />

Mencevax ACWY must be reconstituted by adding the entire contents of the<br />

diluent container to the vial and shaking until the powder is completely<br />

dissolved. Reconstituted vaccine should be used as soon as practicable. If storage<br />

is necessary, hold at +2°C to +8°C for not more than 8 hours.<br />

Menomune must be reconstituted by adding the entire contents of the diluent<br />

container to the vial and shaking until the powder is completely dissolved.<br />

Reconstituted vaccine must be used within 24 hours.<br />

4.10.6 Dosage and administration<br />

Conjugate vaccines<br />

<strong>The</strong> dose of all meningococcal conjugate vaccines (MenCCV, Hib-MenCCV,<br />

4vMenCV) is 0.5 mL to be given by IM injection. Do not mix with other vaccines<br />

in the same syringe.<br />

MenCCVs are registered for use in infants from 6 weeks of age. Hib-MenCCV<br />

is registered for use in infants from 6 weeks of age, and can be administered as<br />

the primary dose of serogroup C meningococcal vaccine and the booster dose<br />

of Hib vaccine.<br />

Both 4vMenCVs may be given from 9 months of age (see 4.10.12 Variations<br />

from product information below).<br />

Either MenCCV or Hib-MenCCV may be administered simultaneously<br />

with other vaccines in the NIP schedule (see 4.10.12 Variations from product<br />

information below).<br />

Polysaccharide vaccines<br />

<strong>The</strong> dose of both 4vMenPVs is 0.5 mL, to be given by SC injection.<br />

4vMenPVs are registered for use in children ≥2 years of age, adolescents and<br />

adults.<br />

4vMenPV may also be co-administered with other vaccines.<br />

288 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Interchangeability of meningococcal vaccines<br />

Experience from the use of conjugate Hib vaccines suggests that the different<br />

brands of conjugate meningococcal vaccines are interchangeable.<br />

<strong>The</strong>re are limited data available on the length of time that should elapse before<br />

administration of 4vMenCV after giving MenCCV. <strong>The</strong> ATAGI recommends a<br />

minimum period of 8 weeks before 4vMenCV is given.<br />

<strong>The</strong>re are limited data available on the length of time that should elapse<br />

before administration of either MenCCV or 4vMenCV after giving 4vMenPV.<br />

A minimum period of 6 months is recommended before conjugate vaccine is<br />

given. 37,39,40<br />

4.10.7 Recommendations<br />

Previous meningococcal disease, regardless of the serogroup, is not a<br />

contraindication to the administration of any meningococcal vaccine.<br />

Meningococcal C conjugate-containing vaccines<br />

Children aged 12 months<br />

It is recommended that a single dose of MenCCV or Hib-MenCCV be given<br />

to all children at the age of 12 months.<br />

Hib-MenCCV is administered as a single dose at 12 months of age where a<br />

booster dose of Hib and the primary dose of serogroup C meningococcal vaccine<br />

are required.<br />

Vaccination before 12 months of age is not recommended, except in infants<br />

with inherited defects of properdin or complement, or functional or anatomical<br />

asplenia (see ‘Persons at high risk for meningococcal disease’ below). 43,44 Infants,<br />

other than those described in the circumstances below, who receive dose(s) of<br />

vaccine at


• Infants from 6 weeks to 12 months of age who have medical conditions<br />

that put them at high risk of meningococcal disease, such as functional or<br />

anatomical asplenia43 or inherited defects of properdin or complement,<br />

persons receiving treatment with eculizumab (a monoclonal antibody<br />

directed against complement component C5), 45 or those post haematopoietic<br />

stem cell transplant (HSCT), are recommended to receive up to 2 doses of<br />

MenCCV vaccine prior to 12 months of age. (See also 3.3 Groups with special<br />

vaccination requirements.)<br />

Infants with these conditions who are 6 weeks to


• Children (aged ≥9 months) and adults with high-risk medical conditions,<br />

such as functional or anatomical asplenia43,44 or complement component<br />

disorders (C5-C9, properdin, factor D or factor H), persons receiving<br />

treatment with eculizumab (a monoclonal antibody directed against<br />

complement component C5), 45 or those post HSCT. In persons with these<br />

risk factors, a 2-dose primary schedule of 4vMenCV is recommended, with<br />

doses given approximately 8 weeks apart. 48 Give 4vMenCV at least 8 weeks<br />

after any previous MenCCV doses. In young children who have received 1 or<br />

more doses of MenCCV prior to 12 months of age, the 1st dose of 4vMenCV<br />

is recommended at 12 months of age. <strong>The</strong> 2nd 4vMenCV dose should be<br />

provided by 18 months of age. (See also 3.3 Groups with special vaccination<br />

requirements and Table 3.3.5 Recommendations for vaccination in persons with<br />

functional or anatomical asplenia.) For persons who have previously received a<br />

dose of 4vMenPV, a booster dose of 4vMenCV, 3 years after the last dose of<br />

4vMenPV, is recommended (see ‘Booster doses or revaccination’ below).<br />

Where 4vMenCV is contraindicated, a single dose of 4vMenPV can be given to<br />

persons aged ≥2 years, unless otherwise indicated.<br />

Booster doses or revaccination<br />

Although the duration of protection following 4vMenCV remains unknown,<br />

persons with medical conditions that place them at high risk of meningococcal<br />

disease (as described above) who have completed a 2-dose primary schedule<br />

of 4vMenCV, should receive 4vMenCV at 5-yearly intervals thereafter, until<br />

further data becomes available. 36,49,50 This includes high-risk infants who have<br />

received MenCCV in infancy, followed by 2 doses of 4vMenCV in the 2nd year<br />

of life. (See also 3.3 Groups with special vaccination requirements and Table 3.3.5<br />

Recommendations for vaccination in persons with functional or anatomical asplenia.)<br />

Persons aged ≥9 months with a medical condition that places them at high risk<br />

of meningococcal disease, and who have previously received 4vMenPV, should<br />

receive a booster dose of 4vMenCV, 3 years after their last dose of 4vMenPV.<br />

<strong>The</strong>reafter, administer 4vMenCV every 5 years.<br />

In persons with other risks for meningococcal disease, such as laboratory<br />

personnel or those travelling to endemic or hyperendemic regions, a dose of<br />

4vMenCV should be administered every 5 years if the risk of meningococcal<br />

exposure is ongoing.<br />

For those aged ≥2 years with underlying risk factors, but in whom 4vMenCV is<br />

contraindicated, ongoing boosting with 4vMenPV is recommended at 5-yearly<br />

intervals unless otherwise indicated.<br />

4.10.8 Pregnancy and breastfeeding<br />

Meningococcal vaccines are not routinely recommended for pregnant or<br />

breastfeeding women, 1,51,52 but can be given where clinically indicated (see 4.10.7<br />

Recommendations above).<br />

PART 4 VACCINE-PREVENTABLE DISEASES 291<br />

4.10 MENINGOCOCCAL DISEASE


Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.10.9 Contraindications<br />

<strong>The</strong> only absolute contraindications to meningococcal vaccines are:<br />

• anaphylaxis following a previous dose of any meningococcal vaccine<br />

• anaphylaxis following any vaccine component.<br />

Previous meningococcal disease, regardless of the serogroup, is not a<br />

contraindication to administration of any meningococcal vaccine.<br />

4.10.10 Adverse events<br />

Common adverse events caused by meningococcal conjugate vaccines are<br />

pain, redness and swelling at the site of injection, fever, irritability, drowsiness,<br />

decreased appetite and headaches. 13,21,23-26,29 <strong>The</strong>re are some age-related<br />

differences in the type of adverse events following vaccination, with systemic<br />

adverse events tending to decrease with increasing age, and local adverse events<br />

tending to increase with increasing age. Headache, anorexia, fever and chills<br />

were more likely to be reported in the adolescent and adult age groups following<br />

administration of 4vMenCV. 26,29 Among recipients of 4vMenCV, rash and nausea<br />

were common. However, serious general adverse events are rare. 26,29<br />

<strong>The</strong> United States Vaccine Adverse Event Reporting System (VAERS) previously<br />

reported a series of cases of Guillain-Barré syndrome (GBS) temporally<br />

associated with the introduction of the 4vMenCV, Menactra. 53,54 <strong>The</strong> likelihood<br />

of coincidentally experiencing GBS after administration of 4vMenCV is expected<br />

to be greater among persons with a history of GBS than among persons with<br />

no history of GBS. Recent safety studies, during which over 2 million doses of<br />

Menactra were administered, found there was no risk of GBS after Menactra<br />

in the general population, and extrapolated these data to conclude that<br />

persons with a history of GBS are not at higher risk than they are after other<br />

vaccines that have no association with GBS. 55,56 It is, therefore, recommended<br />

that 4vMenCV can be administered to persons with previous GBS in whom<br />

vaccination is indicated.<br />

Local adverse events after 4vMenPV include erythema, induration, tenderness,<br />

pain and local axillary lymphadenopathy. However, these reactions are usually<br />

mild and infrequent. Fever and chills occur in approximately 2% of young<br />

children, and may persist for 48 hours or longer, but significant general adverse<br />

events are rare.<br />

4.10.11 Public health management of meningococcal disease<br />

Meningococcal disease is notifiable in all states and territories in Australia.<br />

Further instructions about the public health management of meningococcal<br />

disease, including management of cases of meningococcal disease and their<br />

contacts, should be obtained from state/territory public health authorities (see<br />

292 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Appendix 1 Contact details for <strong>Australian</strong>, state and territory government health<br />

authorities and communicable disease control).<br />

If a diagnosis of meningococcal disease is suspected, the patient should be<br />

immediately given parenteral (usually IM) penicillin and transferred to hospital.<br />

A potential outbreak of meningococcal disease in an institutional or community<br />

setting is a public health emergency needing a rapid response from clinicians<br />

and public health practitioners. <strong>The</strong> decision to control an outbreak with a<br />

vaccination program should be made by the appropriate Public Health Unit,<br />

following the Guidelines for the early clinical and public health management of<br />

meningococcal disease in Australia. 2<br />

4.10.12 Variations from product information<br />

<strong>The</strong> product information for meningococcal C conjugate vaccines states that,<br />

under the age of 12 months, either 2 (NeisVac-C) or 3 (Meningitec and Menjugate<br />

Syringe) doses of vaccine are required. <strong>The</strong> ATAGI recommends instead that<br />

meningococcal C vaccination is routinely not recommended before 12 months of<br />

age (unless specifically indicated).<br />

<strong>The</strong> product information for Meningitec states that an allergic reaction following<br />

a previous dose is a contraindication to further doses. <strong>The</strong> ATAGI recommends<br />

instead that the only contraindication is a history of anaphylaxis to a previous<br />

dose or to any of the vaccine components.<br />

<strong>The</strong> product information for NeisVac-C states that the vaccine should not be<br />

administered with pneumococcal conjugate vaccine, hepatitis B vaccine or<br />

PRP-OMP Haemophilus influenzae type b vaccine unless ‘medically important’.<br />

<strong>The</strong> ATAGI recommends instead that the vaccine may be administered<br />

simultaneously with other vaccines in the NIP. <strong>The</strong>re have been publications<br />

citing the co-administration of MenCCV with other combination vaccines and it<br />

was found to be immunogenic and safe. 57,58<br />

<strong>The</strong> product information for Menactra states that a previous episode of Guillain-<br />

Barré syndrome is a contraindication to vaccination with Menactra. <strong>The</strong> ATAGI<br />

recommends instead that either of the available 4vMenCVs can be administered.<br />

<strong>The</strong> product information for Menactra states that this vaccine is indicated for use<br />

in persons aged 2–55 years. <strong>The</strong> ATAGI recommends instead that Menactra can<br />

be given to persons aged ≥9 months of age.<br />

<strong>The</strong> product information for Menveo states that this vaccine is indicated for use<br />

in persons ≥11 years of age. <strong>The</strong> ATAGI recommends instead that Menveo can be<br />

given to persons aged ≥9 months of age.<br />

<strong>The</strong> product information for all meningococcal vaccines (MenCCV, 4vMenCV<br />

and 4vMenPV) states that there are no data on the use of these vaccines in<br />

lactating women. <strong>The</strong> ATAGI recommends that breastfeeding women can be<br />

vaccinated.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 293<br />

4.10 MENINGOCOCCAL DISEASE


<strong>The</strong> product information for both 4vMenCVs states that vaccine should be<br />

administered as a single dose. <strong>The</strong> ATAGI recommends that these vaccines can be<br />

given in a 2-dose primary schedule to children (aged ≥9 months) and adults with<br />

high-risk medical conditions.<br />

<strong>The</strong> product information for both 4vMenCVs states that the need for, or timing<br />

of, booster doses has not yet been determined. <strong>The</strong> ATAGI recommends that,<br />

until more data become available, individuals with underlying risk factors,<br />

including functional or anatomical asplenia, should continue to receive<br />

4vMenCV at 5-yearly intervals.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

294 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.11 MUMPS<br />

4.11.1 Virology<br />

Mumps is a paramyxovirus, genus Rubulavirus, with a single-stranded RNA<br />

genome. It is rapidly inactivated by heat, formalin, ether, chloroform and light. 1<br />

4.11.2 Clinical features<br />

Mumps is an acute viral illness with an incubation period of 12 to 25 days. 2<br />

Transmission is via respiratory secretions, including aerosol transmission, or by<br />

direct contact with saliva or possibly urine. 2 Asymptomatic infection occurs in<br />

one-third of cases. 3 Symptomatic disease ranges from mild upper respiratory<br />

symptoms to widespread systemic involvement. 3 A high proportion of mumps<br />

infections involve non-specific symptoms including fever, headache, malaise,<br />

myalgia and anorexia. 4 <strong>The</strong> characteristic bilateral, or occasionally unilateral,<br />

parotid swelling occurs in 60 to 70% of clinical cases. 4,5 Maximum infectiousness<br />

occurs between 2 days before onset of illness and 4 days afterwards, but patients<br />

may be infectious from 7 days before parotid swelling to 9 days after. 2 Meningeal<br />

symptoms and signs appear in approximately 10% of cases, but permanent<br />

neurologic sequelae are rare. 2 Mumps encephalitis has been estimated to occur<br />

in 1–2 per 10 000 cases, with a case-fatality rate of around 1.0%. 6 Deafness is<br />

relatively common in mumps meningoencephalitis, although permanent nerve<br />

deafness is rare (1 in 20 000 infections). Orchitis (usually unilateral) has been<br />

reported in up to 15 to 30% of clinical mumps cases in post-pubertal males,<br />

but subsequent sterility is rare. 6 Symptomatic involvement of other glands and<br />

organs has been observed less frequently (pancreatitis, oophoritis, hepatitis,<br />

myocarditis, thyroiditis, mastitis). 1,4<br />

Mumps infection during the first trimester of pregnancy may result in<br />

spontaneous abortion. 3,4 Maternal infection is not associated with an increased<br />

risk of congenital malformation. 3,4<br />

4.11.3 Epidemiology<br />

Mumps is reported worldwide. Prior to universal vaccination, mumps was<br />

primarily a disease of childhood with the peak incidence in the 5–9 years<br />

age group. However, since 2000, peak rates have been reported in older<br />

adolescents and young adults, especially the 20–34 years age group. 7-10<br />

Between 2002 and 2004, mumps notifications were the lowest recorded in<br />

Australia, averaging 0.4 per 100 000. 11 In 2005, notifications increased to<br />

1.2 per 100 000, peaking at 2.7 per 100 000 in 2007, but have since declined to<br />

less than 1 per 100 000 since 2009. 10,11 <strong>The</strong>re have also been recent outbreaks of<br />

mumps in the United States and Europe, where the peak rates of disease have<br />

been in the 18–24 years age group. 12-15<br />

PART 4 VACCINE-PREVENTABLE DISEASES 295<br />

4.11 MUMPS


Similar to measles, persons born in the late 1960s to mid-1980s (especially the<br />

1978–1982 birth cohort) are recognised to be at a greater risk of mumps. Many<br />

missed being vaccinated or acquiring mumps infection as infants (when vaccine<br />

coverage was low and disease incidence was decreasing), and may also have<br />

missed catch-up vaccinations during their school years as part of either the<br />

Measles Control Campaign (which only targeted primary-school-aged children)<br />

or the Young Adult Measles Control Campaign (which did not result in high<br />

coverage). 16,17 During outbreaks, mumps attack rates are lowest in persons who<br />

have received 2 doses of mumps-containing vaccines, as this provides optimal<br />

long-term protection. 5,13 In Australia, over the 11-year period from 1996 to 2006,<br />

mumps was reported as the underlying cause of 5 deaths, all in adults aged over<br />

80 years. 7-10<br />

4.11.4 Vaccines<br />

Monovalent mumps vaccine is not available in Australia. Mumps vaccination<br />

is provided using either measles-mumps-rubella (MMR) or measles-mumpsrubella-varicella<br />

(MMRV) vaccines. Two quadrivalent combination vaccines<br />

containing live attenuated measles, mumps, rubella and varicella viruses<br />

(MMRV) are registered in Australia.<br />

Clinical trials of MMR vaccine indicate 95% mumps seroconversion after a single<br />

dose and up to 100% after a 2nd dose. 4 However, outbreak investigations and<br />

post-marketing studies have reported 1-dose vaccine effectiveness to be between<br />

60 and 90%. 13,18 A Cochrane review reported 1-dose vaccine effectiveness to be<br />

between 69% and 81% for the vaccine containing the Jeryl Lynn mumps strain<br />

and between 70% and 75% for the vaccine containing the Urabe strain. 19 While<br />

protection is greater in 2-dose vaccine recipients, recent outbreaks have reported<br />

mumps in 2-dose vaccine recipients, particularly young adults who received<br />

their vaccines more than 10 years previously. 14,15,20,21<br />

Combination MMRV vaccines have been shown, in clinical trials, to produce<br />

similar rates of seroconversion to all four vaccine components compared with<br />

MMR vaccine and monovalent varicella vaccines administered concomitantly at<br />

separate injection sites. 22-25<br />

See further information on MMR and MMRV vaccines in 4.9 Measles and<br />

4.22 Varicella.<br />

296 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Trivalent measles–mumps–rubella (MMR) vaccines<br />

• M-M-R II – CSL Limited/Merck & Co Inc (live attenuated measles<br />

virus [Enders’ attenuated Edmonston strain], mumps virus [Jeryl<br />

Lynn B level strain] and rubella virus [Wistar RA 27/3 strain]).<br />

Lyophilised pellet in a monodose vial with separate diluent. Each<br />

0.5 mL reconstituted dose contains ≥1000 tissue culture infectious<br />

dose 50% (TCID 50 ) of Enders’ attenuated Edmonston measles<br />

virus, ≥12 500 TCID 50 of the Jeryl Lynn B level mumps virus, and<br />

≥1000 TCID 50 of the Wistar RA 27/3 rubella virus; sorbitol; sucrose;<br />

hydrolysed gelatin; human albumin; fetal bovine serum; neomycin.<br />

• Priorix – GlaxoSmithKline (live attenuated measles virus [Schwarz<br />

strain], mumps virus [RIT 4385 strain, derived from the Jeryl Lynn<br />

strain] and rubella virus [Wistar RA 27/3 strain]). Lyophilised pellet<br />

in a monodose vial with a pre-filled diluent syringe. Each 0.5 mL<br />

reconstituted dose contains ≥10 3.0 cell culture infectious dose 50%<br />

(CCID 50 ) of the Schwarz measles virus, ≥10 3.7 CCID 50 of the RIT 4385<br />

mumps virus, and ≥10 3.0 CCID 50 of the Wistar RA 27/3 rubella virus;<br />

lactose; neomycin; sorbitol; mannitol.<br />

Quadrivalent measles-mumps-rubella-varicella (MMRV) vaccines<br />

• Priorix-tetra – GlaxoSmithKline (live attenuated measles virus<br />

[Schwarz strain], mumps virus [RIT 4385 strain, derived from the<br />

Jeryl Lynn strain], rubella virus [Wistar RA 27/3 strain] and varicellazoster<br />

virus [Oka strain]). Lyophilised pellet in a monodose vial with<br />

a pre-filled diluent syringe. Each 0.5 mL reconstituted dose contains<br />

≥103.0 CCID of the Schwarz measles virus, ≥10 50 4.4 CCID of the RIT<br />

50<br />

4385 mumps virus, ≥103.0 CCID of the Wistar RA 27/3 rubella virus,<br />

50<br />

and ≥103.3 plaque-forming units (PFU) of Oka varicella-zoster virus;<br />

lactose; neomycin; sorbitol; mannitol.<br />

• ProQuad – CSL Limited/Merck & Co Inc (live attenuated measles<br />

virus [Enders’ attenuated Edmonston strain], mumps virus [Jeryl<br />

Lynn B level strain], rubella virus [Wistar RA 27/3 strain] and<br />

varicella-zoster virus [Oka/Merck strain]). Lyophilised powder<br />

in a monodose vial with a pre-filled diluent syringe. Each 0.5 mL<br />

reconstituted dose contains ≥103.0 TCID of Enders’ attenuated<br />

50<br />

Edmonston measles virus, ≥104.3 TCID of the Jeryl Lynn B level<br />

50<br />

mumps virus, ≥103.0 TCID of the Wistar RA 27/3 rubella virus, and<br />

50<br />

≥103.99 PFU of Oka/Merck varicella virus; sucrose; hydrolysed gelatin;<br />

urea; sorbitol; monosodium L-glutamate; human albumin; neomycin;<br />

residual components of MRC-5 cells; bovine serum albumin.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 297<br />

4.11 MUMPS


4.11.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 26 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

Both MMR and MMRV vaccines must be reconstituted by adding the entire<br />

contents of the diluent container to the vial containing the pellet and shaking<br />

until the pellet is completely dissolved.<br />

Reconstituted Priorix (MMR), M-M-R II (MMR) and Priorix-tetra (MMRV)<br />

vaccines should be used as soon as practicable. If storage is necessary, hold at<br />

+2°C to +8°C for not more than 8 hours.<br />

Reconstituted ProQuad (MMRV) vaccine must be used within 30 minutes.<br />

4.11.6 Dosage and administration<br />

<strong>The</strong> dose of Priorix (MMR) vaccine for both children and adults is 0.5 mL to be<br />

given by either SC or IM injection.<br />

<strong>The</strong> dose of M-M-R II (MMR) vaccine for both children and adults is 0.5 mL to be<br />

given by SC injection.<br />

For children


4.11.7 Recommendations<br />

Infants aged


MMRV vaccines are not recommended for use in persons ≥14 years of age, due to<br />

a lack of data on safety and immunogenicity/efficacy in this age group. If a dose<br />

of MMRV vaccine is inadvertently given to an older person, this dose does not<br />

need to be repeated.<br />

For further information on the recommendations for MMR and MMRV vaccines,<br />

see 4.9 Measles and 4.22 Varicella.<br />

Serological testing for immunity to mumps<br />

Serological testing for immunity to mumps (and measles, rubella and varicella) is<br />

not recommended before or after routine administration of the 2-dose childhood<br />

schedule of these vaccines.<br />

However, serological testing for mumps (and measles and rubella) can be<br />

performed in cases where a history of natural immunity or 2 doses of vaccine<br />

administration is uncertain (see ‘Adults and adolescents’ above). Serology is<br />

indicated in special situations, such as pre-pregnancy planning (see also 4.9<br />

Measles, 4.18 Rubella and 4.22 Varicella). Serological tests to investigate immunity<br />

to mumps are generally sensitive at detecting antibody produced by both prior<br />

natural infection and vaccination, although sensitivity varies by assay and<br />

the clinical setting (e.g. time since vaccination). 4 Interpretation of the results<br />

of serological testing may be enhanced by discussion with the laboratory that<br />

performed the test, ensuring that relevant clinical information is provided. An<br />

alternative to serological testing is presumptive administration of MMR vaccine<br />

dose(s). <strong>The</strong>re is no known increase in adverse events from vaccinating those<br />

with pre-existing immunity to one or more of the vaccine components (see 4.11.11<br />

Adverse events below).<br />

4.11.8 Pregnancy and breastfeeding<br />

MMR-containing vaccines are contraindicated in pregnant women. Pregnancy<br />

should be avoided for 28 days after vaccination. 29<br />

MMR vaccines can be given to breastfeeding women. (See also 4.18 Rubella.)<br />

MMRV vaccines are not recommended for use in persons aged ≥14 years.<br />

See also 4.9 Measles, 4.18 Rubella, 4.22 Varicella and 3.3 Groups with special<br />

vaccination requirements, Table 3.3.1 Recommendations for vaccination in pregnancy<br />

for more information.<br />

4.11.9 Contraindications<br />

For information on contraindications to MMR and MMRV vaccines, see 4.9<br />

Measles and 4.22 Varicella.<br />

4.11.10 Precautions<br />

For additional precautions related to MMR and MMRV vaccines, see 4.9 Measles<br />

and 4.22 Varicella.<br />

300 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Vaccination with other live attenuated parenteral vaccines<br />

If MMR or MMRV vaccine is not given simultaneously with other live attenuated<br />

parenteral vaccines (e.g. varicella, BCG, yellow fever), the vaccines should be<br />

given at least 4 weeks apart.<br />

4.11.11 Adverse events<br />

Adverse events following administration of MMR-containing vaccines are<br />

generally mild and well tolerated. 4 Adverse events are much less common after<br />

the 2nd dose of MMR or MMRV vaccine than after the 1st dose.<br />

<strong>The</strong> most common adverse events following mumps vaccination are fever and<br />

parotitis. 4 Parotitis occurs most commonly from 10 to 14 days after vaccination.<br />

<strong>The</strong> incidence varies by vaccine strain; in studies of the Jeryl Lynn vaccine strain,<br />

parotid and/or submandibular swelling occurred in 0.5 to 1.6% of recipients. 4,30,31<br />

An increased risk of aseptic meningitis has been observed after vaccination with<br />

the Urabe strain of mumps vaccine in some countries. 4 However, the Urabe strain<br />

is not used in Australia. MMR and MMRV vaccines available in Australia contain<br />

a Jeryl Lynn-derived strain of mumps, which is not associated with an increased<br />

risk of aseptic meningitis. 32,33<br />

For further information on the adverse events associated with MMR and MMRV<br />

vaccines, see 4.9 Measles and 4.22 Varicella.<br />

4.11.12 Public health management of mumps<br />

Mumps is a notifiable disease in all states and territories in Australia.<br />

Further instructions about the public health management of mumps, including<br />

management of cases of mumps and their contacts, should be obtained from<br />

state/territory public health authorities (see Appendix 1 Contact details for<br />

<strong>Australian</strong>, state and territory government health authorities and communicable<br />

disease control).<br />

Mumps-containing vaccine does not provide protection if given after an<br />

individual has been exposed to mumps. 1,34 However, if the exposure did not<br />

result in infection, the vaccine would induce protection against subsequent<br />

infection. Normal human immunoglobulin (NHIG) has been shown not to be of<br />

value in post-exposure prophylaxis for mumps. 1,34<br />

4.11.13 Variations from product information<br />

For information on MMR and MMRV vaccines, see 4.9 Measles and 4.22 Varicella.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

PART 4 VACCINE-PREVENTABLE DISEASES 301<br />

4.11 MUMPS


4.12 PERTUSSIS<br />

4.12.1 Bacteriology<br />

Pertussis (whooping cough) is caused by Bordetella pertussis, a fastidious, Gramnegative,<br />

pleomorphic bacillus. <strong>The</strong>re are other organisms (such as Bordetella<br />

parapertussis, Mycoplasma pneumoniae and Chlamydia pneumoniae) that can cause a<br />

pertussis-like syndrome. 1<br />

4.12.2 Clinical features<br />

Pertussis is a respiratory infection with an incubation period of 7 to 20 days. In<br />

unvaccinated persons, B. pertussis is highly infectious, spreading by aerosols<br />

to 90% of susceptible household contacts. 2 Natural infection does not provide<br />

long-term protection and repeat infection can occur. 2 <strong>The</strong> characteristic<br />

paroxysmal cough with inspiratory whoop seen in unvaccinated children is less<br />

common in individuals who have varying degrees of immunity acquired from<br />

vaccination or infection. 3 It has been estimated that B. pertussis accounts for up<br />

to 7% of cough illnesses per year in adults and, each year, more than 25% of<br />

adults experience a coughing illness of at least 5 days duration. 4 Even in adults,<br />

pertussis can be associated with significant morbidity, with cough persisting for<br />

up to 3 months, and other significant symptoms, such as sleep disturbance or,<br />

rarely, rib fracture. 5 Identification of pertussis is limited by patient and physician<br />

awareness and, in some cases, the limited sensitivity of diagnostic tests; it is<br />

generally believed to be significantly under-diagnosed (see 4.12.11 Public health<br />

management of pertussis below).<br />

Death due to pertussis is rare in people aged 10–70 years. However, the casefatality<br />

rate in unvaccinated infants


acquired from healthcare workers. 15-18 Pertussis hospitalisation rates for persons<br />

aged ≥60 years are higher than for other adults. 19<br />

Between 2000 and 2010, multiple epidemics of pertussis occurred in Australia;<br />

however, the timing and frequency of these varied by geographical location.<br />

More than 139 000 cases were reported over this 11-year period, with the<br />

highest annual incidence of notifications (156 cases per 100 000 population)<br />

reported in 2010. 20<br />

<strong>The</strong>re have been a number of changes introduced to the NIP schedule over<br />

time in an attempt to improve control of pertussis. Introduction of a 5th dose of<br />

diphtheria, tetanus and whole-cell pertussis vaccine (DTPw) for 4–5-year-old<br />

children in August 1994 was followed by a decrease in notifications consistent<br />

with a vaccine effect; first among children aged 5 and 6 years, then by those<br />

in the 7–9 years age group. 8,21 Subsequently, the average age of pertussis<br />

notifications continued to increase. By 2005, the proportion of notifications in<br />

adults >20 years of age had reached 83%, 8 compared with 40% in the early 1990s.<br />

Acellular pertussis vaccine (DTPa) replaced DTPw for booster doses in 1997,<br />

and for all doses from 1999. In 2003, the DTPa booster dose at 18 months of age<br />

was removed from the NIP, moving the 1st booster dose to 4 years of age. <strong>The</strong><br />

removal of the 18-month booster dose from the schedule was based on evidence<br />

from an Italian longitudinal study of DTPa trial participants. <strong>The</strong> study found<br />

that a primary DTPa course at 2, 4 and 6 months of age provided 76 to 80%<br />

protection from prolonged cough disease and this was maintained until 6 years<br />

of age. 22<br />

In 2009–2010, in contrast to previous epidemics, the highest notification rates<br />

in Australia were in children 20 years of age decreased to 57%. <strong>The</strong> greatest increase in notification<br />

rate occurred in 3-year-old children. Although increased and more sensitive<br />

diagnostic testing using polymerase chain reaction (PCR) has contributed to<br />

this rise, vaccine effectiveness among 3-year olds has been estimated at around<br />

60%, 23 consistent with waning of immunity following the primary DTPa course.<br />

In contrast to notifications, hospitalisation and death rates from pertussis in<br />

the most recent epidemic periods have not increased substantially. 24 A high<br />

proportion of hospitalisations, and almost all deaths, attributed to pertussis<br />

occur in infants too young to have received more than 1 dose of pertussiscontaining<br />

vaccine. 19,25<br />

<strong>The</strong> prevention of severe pertussis morbidity and deaths, particularly in infants<br />


Pregnancy and breastfeeding below). Data to evaluate the effectiveness of indirect<br />

protection to infants from the cocoon approach are lacking. 28 However, this<br />

approach is expected to reduce infection risk to infants from family members,<br />

known to be an important source of pertussis infection, 13,14 especially for the<br />

youngest infants. 11,12<br />

4.12.4 Vaccines<br />

Pertussis vaccine is available in Australia only in combination with diphtheria,<br />

tetanus and other antigens.<br />

<strong>The</strong> acronym DTPa, using capital letters, signifies child formulations of<br />

diphtheria, tetanus and acellular pertussis-containing vaccines. <strong>The</strong> acronym<br />

dTpa is used for formulations that contain substantially lesser amounts<br />

of diphtheria toxoid and pertussis antigens than child (DTPa-containing)<br />

formulations; dTpa vaccines are usually used in adolescents and adults.<br />

Acellular pertussis-containing vaccines have been used for both primary and<br />

booster vaccination of children in Australia since 1999. Whole-cell pertussiscontaining<br />

vaccines were used exclusively before 1997. Between 1997 and 1999<br />

acellular vaccines were used for booster doses. <strong>The</strong>re are a number of acellular<br />

pertussis-containing vaccines that contain three or more purified components<br />

of B. pertussis. In the 3-component vaccines, these components are pertussis<br />

toxin (PT), filamentous haemagglutinin (FHA) and pertactin (PRN). In the<br />

5-component vaccines, fimbrial (FIM) antigens are also included.<br />

Pertussis vaccines provide good protection against severe and typical pertussis,<br />

but substantially less against milder coughing illness. 29,30 Vaccine efficacy of<br />

DTPa vaccines with three or more antigens has been reported as 71 to 78%<br />

for preventing milder symptoms of pertussis and 84% for preventing typical<br />

disease. 30 Epidemiological data suggest that receipt of the 1st dose of the<br />

primary DTPa course significantly reduces the incidence of severe pertussis<br />

disease in young infants, as measured by hospitalisation rates. 31-33 Data on the<br />

duration of immunity following DTPa vaccine indicate that waning occurs 5 to<br />

6 years after the last dose of vaccine. 2,34 However, these studies could not control<br />

for levels of circulating pertussis in the population, which may boost the level<br />

of immunity and lead to over-estimation of the duration of protection against<br />

symptomatic disease. 29,30<br />

Reduced antigen content formulation, dTpa, vaccines are immunogenic. 35-38<br />

A randomised trial in adults reported a point estimate of 92% efficacy against<br />

culture/nucleic acid test-positive disease within 2.5 years of vaccination<br />

with a 3-component monovalent pertussis vaccine. 4 Data on the duration of<br />

immunity to pertussis following a single booster dose of dTpa are limited. Longterm<br />

follow-up of adults vaccinated with dTpa has shown a rapid decline in<br />

levels of pertussis antibodies within the first 2 years after vaccination, with a<br />

continued steady decline out to 10 years after vaccination, although antibody<br />

levels remained above baseline. 39 A similar long-term follow-up of adolescents<br />

304 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


demonstrated a more rapid decline, with pertussis antibody levels decreasing<br />

to or approaching pre-vaccination levels after 10 years. 40 <strong>The</strong> rate of decline<br />

in clinical protection is unknown, but some protection against clinical disease<br />

is likely to persist for up to 10 years. Recent studies have indicated that dTpa<br />

vaccine is immunogenic in the elderly. 38<br />

Vaccines containing DTPa are available in various combinations with inactivated<br />

poliomyelitis, hepatitis B and Haemophilus influenzae type b vaccines.<br />

Formulations for children aged


formaldehyde, glutaraldehyde, polysorbate 80, polymyxin, neomycin<br />

and streptomycin.<br />

• Quadracel – Sanofi Pasteur Pty Ltd (DTPa-IPV; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus). Each 0.5 mL monodose vial<br />

contains ≥30 IU diphtheria toxoid, ≥40 IU tetanus toxoid, 20 µg PT,<br />

20 µg FHA, 3 µg PRN, 5 µg FIM 2+3, 40 D-antigen units inactivated<br />

poliovirus type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and<br />

32 D-antigen units type 3 (Saukett); 1.5 mg aluminium phosphate;<br />

≤50 ng bovine serum albumin; phenoxyethanol as preservative; traces<br />

of formaldehyde, glutaraldehyde, polysorbate 80, polymyxin and<br />

neomycin.<br />

• Tripacel – Sanofi Pasteur Pty Ltd (DTPa; diphtheria-tetanus-acellular<br />

pertussis). Each 0.5 mL monodose vial contains ≥30 IU diphtheria<br />

toxoid, ≥40 IU tetanus toxoid, 10 µg PT, 5 µg FHA, 3 µg PRN, 5 µg<br />

FIM 2+3; 1.5 mg aluminium phosphate; 3.4 mg phenoxyethanol.<br />

Reduced antigen formulations for adults, adolescents and children<br />

aged ≥10 years<br />

• Adacel – Sanofi Pasteur Pty Ltd (dTpa; diphtheria-tetanus-acellular<br />

pertussis). Each 0.5 mL monodose vial contains ≥2 IU diphtheria<br />

toxoid, ≥20 IU tetanus toxoid, 2.5 µg PT, 5 µg FHA, 3 µg PRN,<br />

5 µg FIM 2+3; 0.33 mg aluminium as aluminium phosphate;<br />

phenoxyethanol as preservative; traces of formaldehyde and<br />

glutaraldehyde.<br />

• Adacel Polio – Sanofi Pasteur Pty Ltd (dTpa-IPV; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus). Each 0.5 mL monodose vial<br />

or pre-filled syringe contains ≥2 IU diphtheria toxoid, ≥20 IU tetanus<br />

toxoid, 2.5 µg PT, 5 µg FHA, 3 µg PRN, 5 µg FIM 2+3, 40 D-antigen<br />

units inactivated poliovirus type 1 (Mahoney), 8 D-antigen units type<br />

2 (MEF-1) and 32 D-antigen units type 3 (Saukett); 0.33 mg aluminium<br />

as aluminium phosphate; phenoxyethanol as preservative; traces of<br />

formaldehyde, glutaraldehyde, polysorbate 80, polymyxin, neomycin<br />

and streptomycin.<br />

• Boostrix – GlaxoSmithKline (dTpa; diphtheria-tetanus-acellular<br />

pertussis). Each 0.5 mL monodose vial or pre-filled syringe contains<br />

≥2 IU diphtheria toxoid, ≥20 IU tetanus toxoid, 8 µg PT, 8 µg FHA,<br />

2.5 µg PRN, adsorbed onto 0.5 mg aluminium as aluminium<br />

hydroxide/phosphate; traces of formaldehyde, polysorbate 80 and<br />

glycine.<br />

306 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


• Boostrix-IPV – GlaxoSmithKline (dTpa-IPV; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus). Each 0.5 mL pre-filled<br />

syringe contains ≥2 IU diphtheria toxoid, ≥20 IU tetanus toxoid, 8 µg<br />

PT, 8 µg FHA, 2.5 µg PRN, 40 D-antigen units inactivated poliovirus<br />

type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32 D-antigen<br />

units type 3 (Saukett), adsorbed onto 0.5 mg aluminium as aluminium<br />

hydroxide/phosphate; traces of formaldehyde, polysorbate 80,<br />

polymyxin and neomycin.<br />

4.12.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 41 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

Infanrix hexa must be reconstituted by adding the entire contents of the syringe<br />

to the vial and shaking until the pellet is completely dissolved. Reconstituted<br />

vaccine should be used as soon as practicable. If storage is necessary, hold at<br />

room temperature for not more than 8 hours.<br />

4.12.6 Dosage and administration<br />

<strong>The</strong> dose of all pertussis-containing vaccines is 0.5 mL to be given by IM<br />

injection.<br />

Do not mix DTPa- or dTpa-containing vaccines with any other vaccine in the<br />

same syringe, unless specifically registered for use in this way.<br />

4.12.7 Recommendations<br />

Infants and children<br />

Pertussis-containing vaccine is recommended in a 3-dose primary schedule for<br />

infants at 2, 4 and 6 months of age. Due to the high morbidity and occasional<br />

mortality associated with pertussis in the first few months of life, the 1st dose can<br />

be given as early as 6 weeks of age (see Table 2.1.5 Minimum acceptable age for the<br />

1st dose of scheduled vaccines in infants in special circumstances). Giving a 1st dose at<br />

6 weeks of age rather than 2 months of age is estimated to prevent an additional<br />

8% of infant pertussis cases. <strong>The</strong> next scheduled doses should still be given at<br />

4 months and 6 months of age. 31,42<br />

A booster dose of pertussis-containing vaccine, usually provided as DTPa-IPV,<br />

is recommended at 4 years of age, but can be given as early as 3.5 years of age.<br />

This booster dose is essential as waning of pertussis immunity occurs following<br />

receipt of the primary schedule. 2,34 For this booster dose, all brands of DTPacontaining<br />

vaccines are considered interchangeable.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 307<br />

4.12 PERTUSSIS


Where required, DTPa-containing vaccines can be given for catch-up for either<br />

the primary doses or booster dose in children aged


in the previous 10 years. 19,45 Adults of all ages who require a booster dose of dT<br />

vaccine should be encouraged to do so with dTpa vaccine, particularly if they<br />

have not received a dTpa dose previously (see 4.19 Tetanus and 4.2 Diphtheria). 46<br />

Travellers should receive a booster dose of dT (or dTpa if not given previously)<br />

if more than 10 years have elapsed since the last dose of dT-containing vaccine.<br />

For persons undertaking high-risk travel, consider giving a booster dose of either<br />

dTpa or dT (as appropriate) if more than 5 years have elapsed since the last dose<br />

of a dT-containing vaccine (see 4.19 Tetanus and 4.2 Diphtheria).<br />

For those adults requiring additional protection from polio (see 4.14 Poliomyelitis),<br />

dTpa-IPV can be used.<br />

For additional information on adults with no history of a primary course of dT<br />

or pertussis-containing vaccine requiring catch-up, see 4.19 Tetanus and 2.1.5<br />

Catch-up.<br />

Persons in contact with infants and others at increased risk from pertussis<br />

<strong>The</strong>re is significant morbidity associated with pertussis infection in infants<br />


Healthcare workers<br />

All healthcare workers should receive dTpa vaccine because of the significant<br />

risk of nosocomial transmission of pertussis to vulnerable patients. 15-18 (See<br />

also 3.3 Groups with special vaccination requirements, Table 3.3.7 Recommended<br />

vaccinations for persons at increased risk of certain occupationally acquired vaccinepreventable<br />

diseases.) A booster dose of dTpa is recommended if 10 years have<br />

elapsed since a previous dose. 39,40 Vaccinated healthcare workers who develop<br />

symptoms compatible with pertussis should still be investigated for pertussis.<br />

<strong>The</strong>re have been cases of nosocomial transmission of pertussis to infants from<br />

healthcare workers who have previously received dTpa vaccine. 17<br />

Staff working in early childhood education and care<br />

Adults working with infants and young children aged


(see ‘Women who are planning pregnancy, pregnant or post-partum’ in 4.12.7<br />

Recommendations above).<br />

Recently, it has been recommended in the United States, the United Kingdom and<br />

New Zealand that dTpa vaccine administered in the last trimester of pregnancy<br />

be preferred to post-partum maternal immunisation, for various reasons,<br />

including difficulties in implementation of the latter strategy, theoretical reasons<br />

why vaccination during pregnancy might be more effective, and the favourable<br />

safety profile of maternal vaccination. 53 <strong>The</strong>oretical reasons for effectiveness are:<br />

1) that administration of dTpa during the third trimester of pregnancy results in<br />

high maternal pertussis antibody levels and transplacental transfer of antibodies<br />

to the infant; 54,55 and 2) that the levels of pertussis antibodies measured in cord<br />

blood in such infants are similar to those observed in infants at 12 months of age<br />

following a 3-dose DTPa course. 56 This is likely to provide protection against<br />

pertussis in the 1st month of an infant’s life, prior to commencement of the<br />

primary DTPa schedule. In addition, a woman vaccinated with dTpa during<br />

pregnancy will herself be protected against pertussis during the third trimester<br />

and will be less likely to transmit pertussis to the infant after delivery. Although<br />

specific safety data are limited, dTpa is an inactivated vaccine and there is<br />

evidence for the safety of this and other inactivated vaccines, such as tetanus and<br />

influenza, in pregnancy. 53,57 Safety of dTpa vaccines in pregnancy was not studied<br />

specifically during pre-market evaluations, but review of the available data from<br />

pregnancy registries, small studies and the United States Vaccine Adverse Event<br />

Reporting System did not indicate a higher frequency or unusual pattern of<br />

adverse events in pregnant women who received dTpa. 53<br />

A potential disadvantage of giving dTpa in the third trimester of pregnancy<br />

is that maternal pertussis antibodies may interfere with an infant’s immune<br />

response following the primary 3-dose DTPa course at 2, 4 and 6 months of age, 53<br />

a phenomenon referred to as ‘blunting’. 27,53,58 However, because correlates of<br />

protection are not fully understood, the clinical importance of this is uncertain.<br />

Although severe morbidity and mortality from pertussis is rare after 6 months of<br />

age, it is possible that a reduced immune response to the infant’s primary course<br />

of DTPa-containing vaccine could result in less protection against pertussis in the<br />

2nd year of life. This provides the rationale for provision of an additional booster<br />

dose of a pertussis-containing vaccine (DTPa) in the 2nd year of life to children<br />

born to mothers who received dTpa during pregnancy (see ‘Infants and children’<br />

in 4.12.7 Recommendations above).<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 311<br />

4.12 PERTUSSIS


4.12.9 Contraindications<br />

<strong>The</strong> only absolute contraindications to acellular pertussis-containing<br />

vaccines are:<br />

• anaphylaxis following a previous dose of any acellular<br />

pertussis-containing vaccine<br />

• anaphylaxis following any vaccine component.<br />

4.12.10 Adverse events<br />

Acellular pertussis vaccines are associated with a much lower incidence of fever<br />

(approximately 20%) and local adverse events (approximately 10%) than wholecell<br />

pertussis vaccines (approximately 45% and 40%, respectively), which are no<br />

longer used in Australia. 29,30,59 <strong>The</strong> reduced antigen content dTpa vaccines are safe<br />

and well tolerated in adults. 35,60,61 <strong>The</strong> incidence of fever is low, and comparable<br />

in vaccinated and placebo trial participants. 35,60,61 Booster doses of dTpa within<br />

10 years are also safe and well tolerated in adults. Limb swelling reactions from<br />

booster doses are rare. 39,40,52<br />

Extensive limb swelling, defined as swelling and/or redness involving at least<br />

half the circumference of the limb and the joints both above and below the<br />

injection site, is a recognised adverse event that occurs rarely following booster<br />

doses of DTPa. Such reactions commence within 48 hours of vaccination, last<br />

for 1 to 7 days and resolve completely without sequelae. 43 <strong>The</strong> pathogenesis of<br />

extensive limb swelling is poorly understood. In an analysis of 4th and 5th dose<br />

follow-up studies that examined 12 different DTPa vaccines, 2% of 1015 children<br />

who received consecutive doses of the same DTPa vaccine reported entire thigh<br />

swelling, which resolved completely. 43 A history of extensive limb swelling after<br />

a booster dose of DTPa is not a contraindication to reduced antigen formulations<br />

of dTpa at 11–13 years of age (or older). 62 Parents of children about to receive<br />

the booster dose of a DTPa-containing vaccine (at 4 years of age) should be<br />

informed of the small but well-defined risk of this adverse event which, even<br />

when extensive, is usually not associated with significant pain or limitation of<br />

movement.<br />

Febrile convulsions are very infrequently reported following DTPa-containing<br />

vaccines, within 48 hours of vaccination. <strong>The</strong> risk is even lower in infants who<br />

complete their primary course at 6 months of age, as febrile convulsions are<br />

uncommon in children


an HHE. An HHE may last from a few minutes to 36 hours. In Australia during<br />

2009, 3.2 cases of HHE were reported per 100 000 doses of DTPa-containing<br />

vaccine given to children


To reduce the risk of transmission of B. pertussis, persons with pertussis infection<br />

should commence appropriate antibiotic therapy on clinical suspicion, if within<br />

21 days of the onset of coryza. Antibiotic treatment does not shorten the course<br />

of the illness, but reduces infectivity if provided early in the illness. Detailed<br />

information regarding appropriate macrolide antibiotics and dosing can be found<br />

in the national guidelines for control of pertussis. 72,74<br />

Management of contacts of cases<br />

Vaccination<br />

Since a primary vaccination course requires three or more injections to protect<br />

against pertussis, infant vaccination cannot be effectively used to protect<br />

unimmunised infants. Vaccination has not been shown to have a role in<br />

controlling outbreaks at any age, even in closed settings. However, unvaccinated<br />

or partially vaccinated contacts, up to their <strong>10th</strong> birthday, should be offered<br />

DTPa-containing vaccines, and older contacts should be offered dTpa (see 2.1.5<br />

Catch-up).<br />

Passive immunisation with normal human immunoglobulin is not effective in the<br />

prevention of pertussis.<br />

Chemoprophylaxis<br />

<strong>The</strong> benefit of chemoprophylaxis in preventing the secondary transmission<br />

of pertussis is limited due to multiple factors, including delayed clinical<br />

presentation, delayed diagnosis and imperfect compliance. 75 <strong>The</strong> use of<br />

chemoprophylaxis for prevention of secondary cases should be limited to<br />

close contacts of cases in the household setting who are vulnerable to severe<br />

complications of pertussis, or who, in settings such as early childhood education<br />

and care or healthcare facilities, may transmit pertussis to vulnerable contacts.<br />

Further recommendations regarding chemoprophylaxis of close contacts can be<br />

found in the national guidelines for control of pertussis. 72<br />

4.12.12 Variations from product information<br />

<strong>The</strong> product information for Infanrix hexa states that this vaccine is indicated<br />

for primary immunisation of infants from the age of 6 weeks. <strong>The</strong> ATAGI<br />

recommends that this vaccine may also be used for catch-up of the primary<br />

schedule in children


<strong>The</strong> product information for Quadracel states that this vaccine is indicated for<br />

use in a 3-dose primary schedule from the age of 2 months to 12 months and<br />

may also be used as a booster dose for children from 15 months to 6 years of age<br />

who have previously been vaccinated against diphtheria, tetanus, pertussis and<br />

poliomyelitis. <strong>The</strong> ATAGI recommends that, when appropriate, this product may<br />

also be used for either catch-up of the primary schedule or as a booster dose in<br />

children aged


<strong>The</strong> product information for Boostrix, Boostrix-IPV, Infanrix hexa and<br />

Infanrix IPV states that these vaccines are contraindicated in children with<br />

encephalopathy of unknown aetiology or with neurologic complications<br />

occurring within 7 days following a vaccine dose. <strong>The</strong> ATAGI recommends<br />

instead that the only contraindication is a history of anaphylaxis to a previous<br />

dose or to any of the vaccine components.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

316 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.13 PNEUMOCOCCAL DISEASE<br />

4.13.1 Bacteriology<br />

Streptococcus pneumoniae (pneumococcus) is a Gram-positive coccus. <strong>The</strong><br />

polysaccharide capsule is the most important virulence factor of pneumococci. 1,2<br />

Over 90 capsular antigenic types (serotypes) have been recognised, each of which<br />

elicits type-specific immunity in the host. 3,4 <strong>The</strong> natural reservoir of pneumococci<br />

is the mucosal surface of the upper respiratory tract of humans. 1,3 In a large<br />

majority of hosts, pneumococci are carried with no apparent symptoms. Different<br />

pneumococcal serotypes vary in their propensity to cause nasopharyngeal<br />

colonisation or disease. Worldwide, only a limited number of serotypes are<br />

responsible for most cases of invasive pneumococcal disease (IPD) but the<br />

predominant serotypes vary by age group and geographic area. 5,6 Antibiotic<br />

resistance in pneumococci is an increasing challenge; in 2006, 11% of <strong>Australian</strong><br />

IPD isolates were non-susceptible to penicillin and 3% were non-susceptible to<br />

ceftriaxone/cefotaxime. 7<br />

4.13.2 Clinical features<br />

Person-to-person transmission of S. pneumoniae occurs via contact with<br />

respiratory droplets of colonised persons. Almost all pneumococcal disease<br />

probably begins with the establishment of nasopharyngeal colonisation. From<br />

the nasopharynx, pneumococci may spread locally into adjacent sites to cause<br />

sinusitis, otitis media or pneumonia. Pneumococci may enter the bloodstream,<br />

and also localise in the meninges, causing meningitis, or at other sites including<br />

bones, joints and soft tissues. 1-3,5 For disease surveillance purposes, detection<br />

of S. pneumoniae in a normally sterile site, such as blood, cerebrospinal fluid<br />

or pleural fluid, by culture or nucleic acid testing, is classified as IPD. <strong>The</strong><br />

major clinical categories of IPD are meningitis, bacteraemic pneumonia, and<br />

bacteraemia without focus. In adults, pneumonia with bacteraemia is the most<br />

common manifestation of IPD. Although more difficult to measure for nonbacteraemic<br />

cases, it is estimated that pneumococci account for over one-third of<br />

all community-acquired pneumonia and up to half of hospitalised pneumonia<br />

in adults. 2,8 In children, the most common manifestation is bacteraemia without<br />

focus, accounting for approximately 70% of IPD, followed by pneumonia with<br />

bacteraemia. Meningitis, although least common, is the most severe category of<br />

IPD and has an estimated case-fatality rate of about 30%. 1,2 Acute otitis media<br />

(AOM) is the most common non-invasive manifestation of pneumococcal disease<br />

in children. S. pneumoniae is detected in 28 to 55% of middle ear aspirates from<br />

children with AOM. 2,5,9<br />

Immunocompromised persons who are unable to mount an adequate immune<br />

response to pneumococcal capsular antigens, including those with asplenia,<br />

have the highest risk of IPD. 1,2,4 Household crowding, exposure to cigarette<br />

smoke, childcare attendance, excessive alcohol consumption and certain nonimmunocompromising<br />

chronic medical conditions are also associated with<br />

PART 4 VACCINE-PREVENTABLE DISEASES 317<br />

4.13 PNEUMOCOCCAL DISEASE


greater risk and/or severity of IPD. 1,2,10,11 Indigenous populations in developed<br />

countries, including Aboriginal and Torres Strait Islander people in Australia,<br />

have a disproportionately high burden of IPD. 1,12,13<br />

4.13.3 Epidemiology<br />

<strong>The</strong> highest incidence of IPD is seen at the extremes of age, in young children<br />

and the elderly. 5,7 In Australia, vaccination with 7-valent pneumococcal<br />

conjugate vaccine (7vPCV) was first funded under the NIP from mid-2001, to<br />

5 years of age for Indigenous children living in Central Australia and children<br />

with specified predisposing medical conditions, and to 2 years of age for non-<br />

Indigenous children living in Central Australia and Indigenous children in the<br />

rest of the country. One dose of the 23-valent pneumococcal polysaccharide<br />

vaccine (23vPPV) at 18–24 months of age, as a booster following a primary<br />

7vPCV schedule, was also funded for Indigenous children without predisposing<br />

medical conditions living in jurisdictions with the highest incidence of IPD (the<br />

Northern Territory, Queensland, South Australia and Western Australia). From<br />

January 2005, NIP-funded 7vPCV was extended to all infants nationally, together<br />

with catch-up vaccination for all children aged


non-Indigenous children (88%). 19,20 Since the implementation of the universal<br />

7vPCV program, increased rates of IPD caused by certain serotypes not<br />

contained in 7vPCV (replacement disease) have been observed in Australia and<br />

several other countries. This is particularly so among non-Indigenous children<br />

aged


Pneumococcal conjugate vaccines<br />

• Synflorix – GlaxoSmithKline (10-valent pneumococcal conjugate<br />

vaccine; 10vPCV). Each 0.5 mL monodose vial or pre-filled syringe<br />

contains 1 µg of pneumococcal capsular polysaccharide of serotypes<br />

1, 5, 6B, 7F, 9V, 14, 23F and 3 µg of serotype 4, conjugated to a total of<br />

9–16 µg of non-typeable H. influenzae protein D, 3 µg of serotype 18C<br />

conjugated to 5–10 µg of tetanus toxoid carrier protein, and 3 µg of<br />

serotype 19F conjugated to 3–6 µg of diphtheria toxoid carrier protein,<br />

adsorbed onto 0.5 mg aluminium as aluminium phosphate.<br />

• Prevenar 13 – Pfizer Australia Pty Ltd (13-valent pneumococcal<br />

conjugate vaccine; 13vPCV). Each 0.5 mL monodose pre-filled syringe<br />

contains 2.2 µg each of pneumococcal capsular polysaccharide of<br />

serotypes 1, 3, 4, 5, 6A, 7F, 9V, 14, 18C, 19A, 19F, 23F and 4.4 µg of<br />

serotype 6B, conjugated to non-toxic Corynebacterium diphtheriae<br />

CRM 197 protein, adsorbed onto 0.565 mg aluminium phosphate;<br />

succinic acid; polysorbate 80.<br />

7-valent pneumococcal conjugate vaccine (7vPCV)<br />

A 7vPCV with the mutant non-toxic diphtheria CRM protein as the conjugating<br />

197<br />

protein (Prevenar) became available in Australia in 2001. Efficacy data on the<br />

7vPCV from a pivotal trial in the United States found greater than 95% protective<br />

efficacy against IPD caused by the serotypes contained in the vaccine. 22 A<br />

Cochrane review of clinical trials estimated an efficacy of 80% against vaccinetype<br />

IPD for PCVs (most, but not all, of which used CRM as the conjugating<br />

197<br />

protein) in children


10-valent pneumococcal conjugate vaccine (10vPCV)<br />

10vPCV has been registered for use in Australia since 2009 and is included under<br />

the NIP. <strong>The</strong> protein D of non-typeable H. influenzae (NTHi) is one of the main<br />

conjugating proteins in this vaccine. This vaccine was used for all children aged<br />


of superior effectiveness against IPD or non-IPD pneumonia, the relative benefit<br />

of 13vPCV over 23vPPV for adults is uncertain, since the serotype coverage of<br />

13vPCV is more limited. It is also uncertain whether the level of reduction in<br />

IPD due to the additional serotypes contained in 13vPCV among adults (herd<br />

immunity effect) will be similar to that seen following widespread use of 7vPCV<br />

in children.<br />

Pneumococcal polysaccharide vaccine<br />

• Pneumovax 23 – CSL Limited/Merck & Co Inc (23-valent<br />

pneumococcal polysaccharide vaccine; 23vPPV). Each 0.5 mL<br />

monodose vial contains 25 µg each of pneumococcal capsular<br />

polysaccharide of serotypes 1, 2, 3, 4, 5, 6B, 7F, 8, 9N, 9V, 10A, 11A,<br />

12F, 14, 15B, 17F, 18C, 19F, 19A, 20, 22F, 23F and 33F; 0.25% phenol.<br />

23vPPV contains polysaccharides derived from the 23 most frequent or most<br />

virulent capsular types of S. pneumoniae isolated from sterile fluids in the United<br />

States in the 1970s/early 1980s, with worldwide serotype distribution and<br />

potential cross-reactive serotypes also taken into consideration. 42 <strong>The</strong>se serotypes<br />

are responsible for most IPD cases in adults in Australia. 23vPPV induces<br />

significant immune responses in immunocompetent adults, including the elderly,<br />

with no substantial differences in immune response between older and younger<br />

subjects, but poor responses in the immunocompromised. 43 In children 65 years. 54<br />

<strong>The</strong>re are no studies on the effectiveness of revaccination with 23vPPV for<br />

disease endpoints, although significant and sustained antibody responses<br />

after revaccination are seen in adults, including the elderly. 55-59 Evidence of<br />

322 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


lesser antibody responses to 2nd or subsequent doses of 23vPPV in adults is<br />

variable, 55-58,60 and, even if present, its correlation with clinical effectiveness is<br />

unknown.<br />

4.13.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 61 Store at<br />

+2°C to +8°C. Do not freeze.<br />

10vPCV should be protected from light.<br />

4.13.6 Dosage and administration<br />

<strong>The</strong> dose of pneumococcal conjugate vaccines (10vPCV, 13vPCV) is 0.5 mL, to<br />

be given by IM injection, in the opposite limb to other injectable vaccines, if<br />

possible.<br />

<strong>The</strong> dose of pneumococcal polysaccharide vaccine (23vPPV) is 0.5 mL, to be<br />

given by either IM or SC injection, in the opposite limb to other injectable<br />

vaccines, if possible. <strong>The</strong> IM route is preferred, as a 3-fold greater rate of injection<br />

site reactions is found following administration of 23vPPV by the SC route. 62<br />

However, a vaccine dose administered subcutaneously does not need to be<br />

repeated.<br />

10vPCV (Synflorix) is registered for use in infants and children aged<br />

6 weeks up to 5 years.<br />

13vPCV (Prevenar 13) is registered for use in infants and children aged<br />

6 weeks up to 5 years and adults aged ≥50 years.<br />

23vPPV (Pneumovax 23) is registered for use in children aged ≥2 years<br />

and in adults.<br />

Co-administration with other vaccines<br />

10vPCV may be concurrently administered with other vaccines routinely<br />

used in the infant schedule.<br />

13vPCV may be concurrently administered with other vaccines in the infant<br />

schedule, including inactivated trivalent influenza vaccine. However, parents/<br />

carers of infants or children who are recommended to receive both influenza<br />

vaccine and 13vPCV should be advised of the increased risk of fever following<br />

concomitant administration of these vaccines (see 4.13.10 Precautions below).<br />

Simultaneous administration of Zostavax with pneumococcal polysaccharide<br />

vaccine is not routinely recommended; if possible, the two vaccines should<br />

be given at least 4 weeks apart. However, inadvertent administration of<br />

Zostavax and pneumococcal polysaccharide vaccine at the same time or at an<br />

interval of less than 4 weeks does not require revaccination. One clinical trial<br />

showed reduced immunogenicity of Zostavax in subjects who received both<br />

vaccines concomitantly, compared with those who received Zostavax alone; the<br />

immunogenicity of 23vPPV was not affected by concurrent administration. 63<br />

PART 4 VACCINE-PREVENTABLE DISEASES 323<br />

4.13 PNEUMOCOCCAL DISEASE


However, an observational study from the United States suggests this may not<br />

impact on Zostavax effectiveness. 64-66 (See also 4.24 Zoster.)<br />

Interchangeability of 10vPCV and 13vPCV<br />

<strong>The</strong>re are no available specific data on the interchangeability of 10vPCV and<br />

13vPCV. Although completion of a primary course of pneumococcal conjugate<br />

vaccine with the same formulation is generally preferred, if vaccination with<br />

10vPCV is commenced (e.g. in children born overseas), completion of the course<br />

with 13vPCV is acceptable.<br />

4.13.7 Recommendations<br />

Children aged


Table 4.13.1: Recommendations for pneumococcal vaccination for children<br />

aged


• Table 2.1.11 Catch-up schedule for 13vPCV (Prevenar 13) and 23vPPV<br />

(Pneumovax 23) in children with a medical condition(s) associated with an increased<br />

risk of IPD, presenting at age


Category B: Conditions associated with an increased risk of IPD<br />

• chronic cardiac disease<br />

» particularly cyanotic heart disease or cardiac failure in children<br />

» excluding hypertension only (in adults)<br />

• chronic lung disease, including:<br />

» chronic lung disease in preterm infants<br />

» cystic fibrosis<br />

» severe asthma in adults (requiring frequent hospital visits and use of<br />

multiple medications)<br />

• diabetes mellitus<br />

• Down syndrome<br />

• alcoholism<br />

• chronic liver disease<br />

• preterm birth at 5 years (but not for<br />

those aged


Indigenous children residing in the <strong>Australian</strong> Capital Territory, New South Wales,<br />

Tasmania and Victoria, who do not have any medical condition(s) associated with an<br />

increased risk of IPD, aged


Children aged >5 years to 15 years) who<br />

have an increased risk of IPD, especially in the Northern Territory, where a dose<br />

of 23vPPV is provided to all Indigenous adolescents at approximately 15 years of<br />

age, based on the very high prevalence of conditions associated with an increased<br />

risk of IPD and incidence of IPD in this sub-population (see ‘Adults aged<br />

≥18 years’ below and 3.1 Vaccination for Aboriginal and Torres Strait Islander people).<br />

For children with a medical condition(s) associated with an increased risk of<br />

IPD, further pneumococcal vaccine doses are recommended, as discussed below,<br />

depending on the child’s level of risk.<br />

Those with the highest increased risk of IPD (List 4.13.1, Category A)<br />

Children aged >5 to 5 to


adult 23vPPV dose. Based on current evidence, 13vPCV is not specifically<br />

recommended for children in this age and risk group, regardless of previous<br />

vaccination history.<br />

For children in this age group with a newly identified medical condition(s)<br />

associated with an increased risk of IPD (List 4.13.1, Category B), a single dose<br />

of 23vPPV is recommended at the time of diagnosis. In the rare situation where<br />

a previous dose of 23vPPV has been given (e.g. in Indigenous children in some<br />

jurisdictions), this dose should be given at least 5 years after the previous 23vPPV<br />

dose. <strong>The</strong> next 23vPPV dose should be given approximately 5–10 years after the<br />

1st 23vPPV dose and counted as their 1st adult 23vPPV dose (see ‘Adults with a<br />

condition(s) associated with an increased risk of IPD’ below).<br />

Adults aged ≥18 years<br />

<strong>The</strong> recommendations for use of 23vPPV in adults who do not have a<br />

condition(s) associated with an increased risk of IPD are summarised in Table<br />

4.13.3. Recommendations for the use of 13vPCV and/or 23vPPV in adults with a<br />

condition(s) associated with an increased risk of IPD (List 4.13.1, Category A or<br />

B) are described in the text below.<br />

<strong>The</strong> number of doses recommended depends on age, Indigenous status and the<br />

presence of a condition(s) associated with an increased risk of IPD. Up to 3 doses<br />

(i.e. 2 revaccinations) of 23vPPV in adulthood are recommended, depending on<br />

these factors. This is based on limited data on adverse events and effectiveness,<br />

as well as uncertainty regarding the clinical significance of blunting of antibody<br />

response (immune hyporesponsiveness) following revaccination with 23vPPV,<br />

especially with multiple revaccinations.<br />

For adults, prior childhood doses of 23vPPV that may have been given at either<br />

18–24 months and/or 4–5 years of age should not be counted; that is, they are not<br />

relevant to the recommendations given in Table 4.13.3. In the Northern Territory,<br />

a dose of 23vPPV is provided to all Indigenous adolescents at approximately<br />

15 years of age, based on the very high prevalence of conditions associated with<br />

an increased risk of IPD and incidence of IPD in this population; this dose should<br />

be considered as a dose received in adulthood for the purpose of limiting the<br />

total lifetime number of 23vPPV doses to 3.<br />

Although 13vPCV is registered for use in adults aged ≥50 years, there is currently<br />

insufficient evidence to recommend its use in preference to 23vPPV at the<br />

individual or population level for persons aged ≥18 years who do not have a<br />

condition(s) associated with an increased risk of IPD (see 4.13.4 Vaccines above).<br />

Updated recommendations on the use of 13vPCV in adults without an increased<br />

risk of IPD will be made when more data are available (see Immunise Australia<br />

website www.immunise.health.gov.au).<br />

330 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Non-Indigenous adults<br />

A single dose of 23vPPV is recommended for adults at 65 years of age. For those<br />

aged >65 years who did not receive a dose at 65 years of age, a single catch-up<br />

dose of 23vPPV should be offered as soon as possible. Routine revaccination with<br />

23vPPV for non-Indigenous adults without a condition(s) associated with an<br />

increased risk of IPD is not recommended (see Table 4.13.3).<br />

Aboriginal and Torres Strait Islander (Indigenous) adults<br />

A 1st dose of 23vPPV is recommended for all Indigenous adults reaching the age<br />

of 50 years (Table 4.13.3). This is based on the increased risk of IPD in Indigenous<br />

adults compared with non-Indigenous adults, and the high prevalence of<br />

conditions associated with an increased risk of IPD (including tobacco smoking)<br />

in Indigenous adults after 50 years of age, compared with younger ages. A 2nd<br />

dose of 23vPPV is recommended 5 years after the 1st dose. For those aged<br />

≥50 years who have never received a dose of 23vPPV, a 1st dose should be<br />

offered as soon as possible, with a 2nd dose recommended 5 years after the<br />

1st dose.<br />

Indigenous adults aged


Table 4.13.3: Recommendations for pneumococcal vaccination using 23vPPV<br />

for adults who do not have a condition(s) associated with an<br />

increased risk of invasive pneumococcal disease (IPD)*<br />

Non-Indigenous adults<br />

Current age (years) 1st dose of 23vPPV 2nd dose of<br />

23vPPV<br />

(1st revaccination)<br />

332 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

3rd dose of 23vPPV<br />

(2nd revaccination)<br />

18 to


Adults who have a condition listed in Category B in List 4.13.1 are not<br />

recommended to receive 13vPCV. 13vPCV is registered for use in children up<br />

to the age of 5 years only and in adults aged ≥50 years, and data in adults and<br />

in immunocompromised persons is limited. However, based on extrapolation<br />

from data on the 7vPCV, 76 providing a dose of 13vPCV to adults at the highest<br />

increased risk of IPD is likely to be beneficial.<br />

Use of 23vPPV<br />

All adults with a condition(s) associated with an increased risk of IPD (List 4.13.1,<br />

Categories A and B) are recommended to receive additional doses of 23vPPV<br />

(compared with those who do not have an increased risk).<br />

In adults with a pre-existing condition (List 4.13.1, Categories A and B) the 1st<br />

adult dose of 23vPPV is recommended at approximately 18 years of age (see also<br />

‘Children aged >5 years to


4.13.8 Pregnancy and breastfeeding<br />

23vPPV is not routinely recommended for pregnant or breastfeeding women.<br />

Although 23vPPV has been administered in pregnancy in the context of<br />

clinical trials77 with no evidence of adverse effects, data are limited; deferral of<br />

vaccination with 23vPPV until after delivery is recommended unless there is<br />

an increased risk of IPD. Women of child-bearing age who have a condition(s)<br />

associated with an increased risk of IPD should be vaccinated before a planned<br />

pregnancy, or as soon as practicable after delivery (see 4.13.7 Recommendations<br />

above). 23vPPV may be given to breastfeeding women.<br />

Data on use the use of 10vPCV and 13vPCV during pregnancy or lactation are<br />

not available. For adults with a condition(s) associated with an increased risk of<br />

IPD for whom a dose of 13vPCV is recommended, the dose should be deferred<br />

until after delivery and cessation of breastfeeding.<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.13.9 Contraindications<br />

<strong>The</strong> only absolute contraindications to pneumococcal vaccines are:<br />

• anaphylaxis following a previous dose of any pneumococcal vaccine<br />

• anaphylaxis following any vaccine component.<br />

4.13.10 Precautions<br />

13-valent pneumococcal conjugate vaccine and inactivated influenza vaccines<br />

One study in the United States has suggested that there is a slightly higher<br />

risk of febrile seizure associated with concurrent administration of 13vPCV<br />

and inactivated trivalent influenza vaccine than after receipt of either of the<br />

vaccines alone on separate days. 67 <strong>The</strong> increased likelihood of febrile seizures<br />

occurred within 1 day of vaccination in children aged 6–59 months who<br />

received 13vPCV concurrently with a 1st dose of inactivated trivalent influenza<br />

vaccine. <strong>The</strong> risk was estimated to be about 18 excess cases per 100 000 doses<br />

for those aged 6–59 months, with a peak of 45 per 100 000 doses for those<br />

aged 16 months. Given this relatively low increase in risk, providing 13vPCV<br />

and inactivated trivalent influenza vaccine concurrently to children aged<br />

12–23 months is acceptable; however, immunisation service providers should<br />

advise parents regarding this, and provide the option of administering these<br />

two vaccines on separate days (with an interval of not less than 3 days). (See<br />

also 4.7 Influenza.) In the event that fever occurs following either vaccine, an<br />

interval may minimise the risk of increased adverse reactions.<br />

334 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.13.11 Adverse events<br />

10-valent pneumococcal conjugate vaccine<br />

<strong>The</strong> safety profile of 10vPCV is similar to that of 7vPCV, 68 with no clinically<br />

relevant difference when co-administered with routine childhood vaccines. 69<br />

In clinical trials, erythema, pain, or swelling of any degree at the injection site<br />

each occurred in approximately 30 to 50% of 10vPCV recipients. Erythema<br />

of >30 mm occurred in up to about 5% of 10vPCV recipients in the primary<br />

course. <strong>The</strong> frequency of local adverse events was higher after the booster<br />

dose. Irritability and drowsiness were reported in about 50% of 10vPCV<br />

recipients when co-administered with a DTPa-combination vaccine, but severe<br />

reactions occurred in fewer than 5%. When co-administered with DTPa-based<br />

combination vaccines, fever with temperature ≥38°C was reported in about 33%<br />

of 10vPCV recipients after primary or booster doses. Approximately 2 to 6% of<br />

10vPCV recipients reported rectal temperature >39°C after primary vaccination<br />

and 1.5 to 3% after booster vaccination. 68<br />

13-valent pneumococcal conjugate vaccine<br />

Pooled safety analysis from 13 clinical trials showed that the safety profile of<br />

13vPCV in young children is similar to that of 7vPCV. 70,78 Pain/tenderness and<br />

erythema at the injection site occurred in about 50% of all 13vPCV recipients, and<br />

induration or swelling in about 33%. Pain interfering with movement occurred<br />

in about 8%. Moderate erythema and induration occurred more commonly after<br />

the toddler dose than after an infant dose, in about 13% of recipients. About<br />

37% of 13vPCV childhood recipients reported fever, with about 5% reporting<br />

fever >39°C. 78 Fever occurred more frequently after the toddler dose than after<br />

the primary doses. 79 Other common systemic reactions included irritability,<br />

drowsiness/increased sleep and decreased appetite, reported in 70%, 60% and<br />

39% of 13vPCV recipients, respectively. 78 Frequencies of each of these adverse<br />

events were comparable to those in 7vPCV recipients. 78<br />

Post-marketing surveillance in the United States has suggested the possibility<br />

of a higher risk of febrile seizure within a day of vaccination among those who<br />

received concurrent administration of 13vPCV and inactivated trivalent influenza<br />

vaccine in 2010–2011, compared with those who received either vaccine alone,<br />

especially in children aged 12–23 months (see 4.13.10 Precautions above). 67<br />

<strong>The</strong>re is only limited information available on the safety of 13vPCV use in<br />

adults, from a study in which adults aged ≥65 years received 13vPCV with or<br />

without concurrent vaccination with the trivalent seasonal influenza vaccine. 80<br />

Pain, redness and/or swelling at the 13vPCV injection site were very common,<br />

occurring in 47% in both groups. <strong>The</strong> common systemic reactions that were<br />

reported significantly more often among 13vPCV than placebo recipients, both<br />

concurrently receiving the trivalent influenza vaccine, were chills, rash and new<br />

muscle pain. Overall, there were no significant differences in adverse event<br />

PART 4 VACCINE-PREVENTABLE DISEASES 335<br />

4.13 PNEUMOCOCCAL DISEASE


frequencies between those with or without prior 23vPPV doses. Recipients of<br />

concurrent administration of inactivated trivalent seasonal influenza vaccine<br />

with 13vPCV reported a higher frequency of systemic but not local reactions. 80<br />

23-valent pneumococcal polysaccharide vaccine<br />

<strong>The</strong> proportion of vaccine recipients reporting local and systemic reactions after a<br />

primary or a repeat dose of pneumococcal polysaccharide vaccines varies among<br />

different study populations, and possibly with age. 55,57,81 About 50% or more of<br />

23vPPV recipients will experience some soreness after the 1st dose, and swelling<br />

and redness are also very common, occurring in approximately 20% of recipients.<br />

Moderate or severe local adverse events that limit arm movement are also quite<br />

common, occurring in up to 5% of 1st dose recipients. Systemic reactions like<br />

myalgia, fatigue and chills are also very common. Fever ≥37.5°C occurs in up to<br />

10% of 23vPPV recipients, but high fever is uncommon. 55,57,81<br />

Larger and more recent studies indicate that both local and systemic adverse<br />

events occur more commonly after a repeat dose of 23vPPV than after the<br />

1st dose in adults, particularly more severe local adverse events, which may<br />

occur in up to approximately 20% of revaccinated subjects. 55,57,81 <strong>The</strong>se findings<br />

effectively supersede the inconsistent findings from some smaller studies, which<br />

were limited by subject numbers and methodology. 82-86 Nevertheless, the local<br />

adverse events were mostly non-serious and self-limiting. In these studies, the<br />

repeat doses were given at least 5 years after the previous dose. Another study,<br />

which used hospitalisations coded as cellulitis or abscess of the upper limb<br />

within 3 days of pneumococcal vaccination as a proxy measure for very severe<br />

local adverse events, showed that these adverse events were significantly more<br />

likely when a repeat dose was given within 5 years of the 1st dose. 87 As severe<br />

local reactions are also associated with higher antibody levels, 57,62,81,88 this is the<br />

likely driver of the relationship with shorter intervals between the repeat and<br />

the primary dose and suggests that such local reactions are associated with more<br />

robust immunity.<br />

4.13.12 Variations from product information<br />

<strong>The</strong> product information for Prevenar 13 recommends 4 doses of 13vPCV for<br />

vaccination commencing at 6 weeks of age, with further doses at 4, 6 and<br />

12–15 months of age; 3 doses for vaccination commencing between 7 and<br />

11 months of age; and 2 doses for vaccination commencing between 12 and<br />

23 months of age. <strong>The</strong> ATAGI recommends instead that 1 dose less than that<br />

stated in the product information be given to healthy children who are not at<br />

increased risk of IPD. <strong>The</strong> ATAGI recommends that the 1st dose be given at<br />

2 months of age, and that this dose can be given as early as 6 weeks of age.<br />

<strong>The</strong> next scheduled doses should be given at 4 months and 6 months of age.<br />

13vPCV is registered for use in children up to 5 years of age and adults aged<br />

≥50 years. <strong>The</strong> ATAGI recommends a dose of 13vPCV for children aged<br />

336 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


5 years with a condition(s) associated with the highest risk of IPD (see List<br />

4.13.1, Category A) who have not completed a primary course of 13vPCV, or<br />

who have not received a supplementary dose of 13vPCV before (see 4.13.7<br />

Recommendations above), or for adults of any age who have a condition(s)<br />

associated with the highest risk of IPD. This is based on the likely benefit<br />

outweighing uncertainties and risks, and on immunogenicity and safety data in<br />

younger children.<br />

<strong>The</strong> product information for Pneumovax 23 states that Pneumovax 23 and<br />

Zostavax should not be given concurrently. <strong>The</strong> ATAGI instead recommends that<br />

while concurrent administration of Zostavax with pneumococcal polysaccharide<br />

vaccine is not routinely recommended, and if possible the two vaccines should<br />

be given at least 4 weeks apart, inadvertent administration of Zostavax and<br />

pneumococcal polysaccharide vaccine at the same time or at an interval of less<br />

than 4 weeks does not require revaccination.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

PART 4 VACCINE-PREVENTABLE DISEASES 337<br />

4.13 PNEUMOCOCCAL DISEASE


4.14 POLIOMYELITIS<br />

4.14.1 Virology<br />

Polioviruses are classified as enteroviruses in the family Picornaviridae. 1 <strong>The</strong>y<br />

have an RNA genome, and can inhabit the gastrointestinal tract transiently.<br />

<strong>The</strong>re are three poliovirus serotypes, referred to as either type 1, type 2 or type 3.<br />

<strong>The</strong> virus enters through the mouth, multiplies in the pharynx and gut and is<br />

excreted in the stools for several weeks. <strong>The</strong> virus invades local lymphoid tissue,<br />

enters the bloodstream and may then infect and replicate in cells of the central<br />

nervous system. 2<br />

4.14.2 Clinical features<br />

Poliomyelitis is an acute illness following gastrointestinal infection by one of the<br />

three types of poliovirus. Transmission is through faecal–oral and, occasionally,<br />

oral–oral spread. 3 <strong>The</strong> infection may be clinically inapparent. If symptoms<br />

occur, they may include headache, gastrointestinal disturbance, malaise and<br />

stiffness of the neck and back, with or without paralysis. Paralysis is classically<br />

asymmetrical. Paralytic polio is a complication of poliovirus aseptic meningitis,<br />

and may be spinal (79%), bulbar (2%) or bulbospinal (19%). <strong>The</strong> case-fatality<br />

rate in paralytic polio is 2 to 5% in children and 15 to 30% in adults. <strong>The</strong> casefatality<br />

rate in bulbar polio is up to 75%. <strong>The</strong> infection rate in households<br />

with susceptible young children can reach 100%. <strong>The</strong> ratio of inapparent or<br />

asymptomatic infection to paralytic infection may be as high as 1000:1 in<br />

children and 75:1 in adults, depending on the poliovirus type and social and<br />

environmental conditions. 2<br />

<strong>The</strong> incubation period ranges from 3 to 21 days. Infected persons are most<br />

infectious from 7 to 10 days before to 7 to 10 days after the onset of symptoms.<br />

<strong>The</strong> oral vaccine virus may be shed in the faeces for 6 weeks or more, 2 and for up<br />

to several years in people who are immunocompromised. Oral vaccine strains<br />

shed for many years may mutate into potentially neurovirulent strains. 4-9<br />

4.14.3 Epidemiology<br />

<strong>The</strong> incidence of poliomyelitis has been dramatically reduced worldwide with<br />

the World Health Organization (WHO) aiming to achieve cessation of all wild<br />

poliovirus transmission worldwide by <strong>2013</strong> through an intensified Global Polio<br />

Eradication Initiative. <strong>The</strong>re have been imported poliomyelitis case reports in<br />

parts of Southeast Asia, eastern Europe and Africa. 10-12 Further information is<br />

available from the WHO Polio Eradication website (www.polioeradication.org).<br />

In 1994, the continents of North and South America were certified to be free of<br />

polio, 13 followed by the Western Pacific region (including Australia) in 200014 and the European region in 2002. 15 In countries where the disease incidence is<br />

low but transmission is still occurring, poliomyelitis cases are seen sporadically<br />

or as outbreaks among non-vaccinated persons. In 2012, polio had been<br />

338 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


virtually eradicated in India, but there were still cases in Afghanistan, Nigeria<br />

and Pakistan. 16<br />

In Australia, the peak incidence of poliomyelitis was 39.1/100 000 in 1938. <strong>The</strong>re<br />

has been a dramatic fall in incidence since 1952, but epidemics occurred in 1956<br />

and 1961–62. <strong>The</strong> most recent laboratory-confirmed case of wild poliomyelitis<br />

in Australia occurred in 2007 in an overseas-born student who acquired the<br />

disease during a visit to Pakistan. 17 <strong>The</strong> last case of wild poliomyelitis prior to<br />

this was in 1977, due to an importation from Turkey, but two vaccine-associated<br />

cases were notified in 1986 and 1995. 18,19 Because of the rapid progress in global<br />

polio eradication and the diminished risk of wild virus-associated disease,<br />

inactivated poliomyelitis vaccine (IPV) is now used for all doses of polio vaccine<br />

in Australia. 3,20 <strong>The</strong> advantage of using IPV is that it cannot cause vaccineassociated<br />

paralytic poliomyelitis (VAPP). 21 Emergence of highly evolved<br />

vaccine-derived polioviruses (VDPV) in persons with primary immunodeficiency<br />

(iVDPV) with long-term excretion, and polio outbreaks due to circulating VDPV<br />

(cVDPV), particularly in areas with low vaccine coverage, are associated with<br />

oral poliomyelitis vaccine (OPV) administration. 22,23<br />

4.14.4 Vaccines<br />

Inactivated poliomyelitis vaccine<br />

• IPOL – Sanofi Pasteur Pty Ltd (IPV; inactivated poliovirus). Each<br />

0.5 mL pre-filled syringe contains 40 D-antigen units inactivated<br />

poliovirus type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and<br />

32 D-antigen units type 3 (Saukett); 2–3 µL phenoxyethanol; 2–20 µg<br />

formaldehyde; polysorbate 80; traces of neomycin, streptomycin,<br />

polymyxin B and bovine serum albumin.<br />

Combination vaccines that contain IPV<br />

Formulations for children aged


polysaccharide (PRP) conjugated to 20–40 µg tetanus toxoid. May<br />

contain yeast proteins.<br />

• Infanrix IPV – GlaxoSmithKline (DTPa-IPV; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus). Each 0.5 mL pre-filled<br />

syringe contains ≥30 IU diphtheria toxoid, ≥40 IU tetanus toxoid,<br />

25 µg PT, 25 µg FHA, 8 µg PRN, 40 D-antigen units inactivated<br />

poliovirus type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1)<br />

and 32 D-antigen units type 3 (Saukett), adsorbed onto aluminium<br />

hydroxide; traces of formaldehyde, polysorbate 80, polymyxin and<br />

neomycin.<br />

• Pediacel – Sanofi Pasteur Pty Ltd (DTPa-IPV-Hib; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus-Haemophilus influenzae type<br />

b). Each 0.5 mL monodose vial contains ≥30 IU diphtheria toxoid,<br />

≥40 IU tetanus toxoid, 20 µg PT, 20 µg FHA, 3 µg PRN, 5 µg pertussis<br />

fimbriae (FIM) 2+3, 40 D-antigen units inactivated poliovirus type<br />

1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32 D-antigen<br />

units type 3 (Saukett), 10 µg Hib capsular polysaccharide conjugated<br />

to 20 µg tetanus protein; 1.5 mg aluminium phosphate; ≤50 ng<br />

bovine serum albumin; phenoxyethanol as preservative; traces of<br />

formaldehyde, glutaraldehyde, polysorbate 80, polymyxin, neomycin<br />

and streptomycin.<br />

• Quadracel – Sanofi Pasteur Pty Ltd (DTPa-IPV; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus). Each 0.5 mL monodose vial<br />

contains ≥30 IU diphtheria toxoid, ≥40 IU tetanus toxoid, 20 µg PT,<br />

20 µg FHA, 3 µg PRN, 5 µg FIM 2+3, 40 D-antigen units inactivated<br />

poliovirus type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and<br />

32 D-antigen units type 3 (Saukett); 1.5 mg aluminium phosphate;<br />

≤50 ng bovine serum albumin; phenoxyethanol as preservative; traces<br />

of formaldehyde, glutaraldehyde, polysorbate 80, polymyxin and<br />

neomycin.<br />

Reduced antigen formulations for adults, adolescents and children aged ≥10 years<br />

• Adacel Polio – Sanofi Pasteur Pty Ltd (dTpa-IPV; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus). Each 0.5 mL monodose vial<br />

or pre-filled syringe contains ≥2 IU diphtheria toxoid, ≥20 IU tetanus<br />

toxoid, 2.5 µg PT, 5 µg FHA, 3 µg PRN, 5 µg FIM 2+3, 40 D-antigen<br />

units inactivated poliovirus type 1 (Mahoney), 8 D-antigen units type<br />

2 (MEF-1) and 32 D-antigen units type 3 (Saukett); 0.33 mg aluminium<br />

as aluminium phosphate; phenoxyethanol as preservative; traces of<br />

340 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


formaldehyde, glutaraldehyde, polysorbate 80, polymyxin, neomycin<br />

and streptomycin.<br />

• Boostrix-IPV – GlaxoSmithKline (dTpa-IPV; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus). Each 0.5 mL pre-filled<br />

syringe contains ≥2 IU diphtheria toxoid, ≥20 IU tetanus toxoid, 8 µg<br />

PT, 8 µg FHA, 2.5 µg PRN, 40 D-antigen units inactivated poliovirus<br />

type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32 D-antigen<br />

units type 3 (Saukett), adsorbed onto 0.5 mg aluminium as aluminium<br />

hydroxide/phosphate; traces of formaldehyde, polysorbate 80,<br />

polymyxin and neomycin.<br />

IPV (IPOL) and IPV-containing combination vaccines contain polioviruses<br />

of types 1, 2 and 3 inactivated by formaldehyde. A course of 3 doses with an<br />

interval of 2 months between each dose produces long-lasting immunity (both<br />

mucosal and humoral) to all three poliovirus types. IPV produces considerably<br />

lower levels of intestinal immunity than OPV.<br />

4.14.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 24 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

Infanrix hexa must be reconstituted by adding the entire contents of the syringe<br />

to the vial and shaking until the pellet is completely dissolved. Reconstituted<br />

vaccine should be used as soon as practicable. If storage is necessary, hold at<br />

room temperature for not more than 8 hours.<br />

4.14.6 Dosage and administration<br />

<strong>The</strong> dose of IPV (IPOL) is 0.5 mL, to be given by SC injection. If IPV (IPOL) is<br />

inadvertently given intramuscularly, there is no need to repeat the dose.<br />

<strong>The</strong> dose of the IPV-containing combination vaccines is 0.5 mL, to be given by<br />

IM injection.<br />

<strong>The</strong> primary course consists of 3 doses of vaccine. An interval of 2 months<br />

between doses is recommended, but the minimum interval can be as short as<br />

1 month for catch-up in children or adults.<br />

Interchangeability of oral and inactivated poliomyelitis vaccine<br />

OPV is no longer in use in Australia. OPV and IPV are interchangeable.<br />

Children commenced on OPV should complete their polio vaccination schedule<br />

using IPV (IPOL) or IPV-containing vaccines. 25<br />

PART 4 VACCINE-PREVENTABLE DISEASES 341<br />

4.14 POLIOMYELITIS


4.14.7 Recommendations<br />

Primary vaccination of infants and children<br />

IPV (IPOL) or IPV-containing vaccines are recommended for infants at 2, 4 and<br />

6 months of age. <strong>The</strong> 1st dose of an IPV-containing vaccine can be given as<br />

early as 6 weeks of age. If the 1st dose is given at 6 weeks of age, the next<br />

scheduled doses should still be given at 4 months and 6 months of age. An<br />

open, randomised, multi-centre trial comparing the hexavalent and pentavalent<br />

IPV-containing vaccines found that infants receiving either vaccine at 2, 4 and<br />

6 months of age had seroprotective levels of antibody to poliovirus types 1, 2<br />

and 3. 26 Extra doses of IPV (IPOL) or IPV-containing vaccines are not needed for<br />

babies born prematurely.<br />

Where only IPV vaccination is required, IPOL can be used for catch-up in<br />

children. If other antigens including poliomyelitis are required, Infanrix IPV<br />

or Infanrix hexa can be used for catch-up in children aged


4.14.8 Pregnancy and breastfeeding<br />

IPV (IPOL) or IPV-containing vaccines are not routinely recommended for<br />

pregnant or breastfeeding women, but can be given where vaccination is<br />

considered necessary (see 4.14.7 Recommendations above).<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.14.9 Contraindications<br />

<strong>The</strong> only absolute contraindications to IPV (IPOL) or IPV-containing vaccines are:<br />

• anaphylaxis following a previous dose of any IPV-containing vaccine<br />

• anaphylaxis following any vaccine component.<br />

4.14.10 Adverse events<br />

IPV (IPOL) or IPV-containing vaccines cause erythema (in 33% of vaccine<br />

recipients), pain (in 13%), and induration (in 1%) at the injection site. Other<br />

symptoms reported in young babies are fever, crying and decreased appetite (in<br />

5 to 10%).<br />

Repeat doses of IPV or IPV-containing vaccines have not been associated with<br />

increased adverse events and, where extra doses are required, are safe.<br />

4.14.11 Public health management of poliomyelitis<br />

Poliomyelitis is a notifiable disease in all states and territories in Australia.<br />

Further instructions about the public health management of poliomyelitis,<br />

including management of cases of poliomyelitis and their contacts, should<br />

be obtained from state/territory public health authorities (see Appendix 1<br />

Contact details for <strong>Australian</strong>, state and territory government health authorities and<br />

communicable disease control).<br />

4.14.12 Variations from product information<br />

<strong>The</strong> product information for Infanrix hexa states that this vaccine is indicated<br />

for primary immunisation of infants from the age of 6 weeks. <strong>The</strong> ATAGI<br />

recommends that this vaccine may also be used for catch-up of the primary<br />

schedule in children


<strong>The</strong> product information for Quadracel states that this vaccine is indicated for<br />

use in a 3-dose primary schedule from the age of 2 months to 12 months and<br />

may also be used as a booster dose for children from 15 months to 6 years of age<br />

who have previously been vaccinated against diphtheria, tetanus, pertussis and<br />

poliomyelitis. <strong>The</strong> ATAGI recommends that, when appropriate, this product may<br />

also be used for either catch-up of the primary schedule or as a booster dose in<br />

children aged


4.15 Q FEVER<br />

4.15.1 Bacteriology<br />

Q fever is caused by Coxiella burnetii, an obligate intracellular bacterium. 1 <strong>The</strong><br />

organism is inactivated at pasteurisation temperatures. It survives well in air,<br />

soil, water and dust, and may also be disseminated on fomites such as wool,<br />

hides, clothing, straw and packing materials. 2,3 C. burnetii has been weaponised<br />

and is considered a Category B biothreat agent. 4<br />

4.15.2 Clinical features<br />

Q fever can be acute or chronic, and there is increasing recognition of long-term<br />

sequelae. Infection is asymptomatic in at least half of cases. 5,6<br />

Acute Q fever usually has an incubation period of 2 to 3½ weeks, depending on<br />

the inoculum size and other variables7 (range from 4 days up to 6 weeks). Clinical<br />

symptoms vary by country, but, in Australia, the most common presentation is<br />

rapid onset of high fever, rigors, profuse sweats, extreme fatigue, muscle and<br />

joint pain, severe headache and photophobia. 5,6 As the attack progresses, there<br />

is usually evidence of hepatitis, occasionally with frank jaundice; a proportion<br />

of patients may have pneumonia, which is usually mild but can require<br />

mechanical ventilation. If untreated, the acute illness lasts 1 to 3 weeks and may<br />

be accompanied by substantial weight loss in more severe cases. 5,6 Infection often<br />

results in time off work, lasting a few days to several weeks. 8<br />

C. burnetii may cause chronic manifestations, the most commonly<br />

reported being subacute endocarditis. Less common presentations include<br />

granulomatous lesions in bone, joints, liver, lung, testis and soft tissues.<br />

Infection in early pregnancy, or even before conception, may recrudesce at term<br />

and cause fetal damage. 9-11<br />

Studies have also identified a late sequela to infection, post Q fever fatigue<br />

syndrome (QFS), which occurs in about 10 to 15% of patients who have<br />

previously had acute Q fever. 12-15 Research suggests that non-infective antigenic<br />

complexes of C. burnetii persist for many years after acute Q fever, and the<br />

maintenance of immune responses to these antigens might be the biological basis<br />

by which QFS occurs. 13,16-18<br />

4.15.3 Epidemiology<br />

C. burnetii infects both wild and domestic animals and their ticks, with cattle,<br />

sheep and goats being the main sources of human infection. 19-21 Companion<br />

animals such as cats and dogs may also be infected, as well as native <strong>Australian</strong><br />

animals such as kangaroos, and introduced animals such as feral cats and<br />

camels. 19,21-23 <strong>The</strong> animals shed C. burnetii into the environment through their<br />

placental tissue or birth fluids, which contain exceptionally high numbers of<br />

Coxiella organisms, and also via their milk, urine and faeces. C. burnetii is highly<br />

infectious24 and can survive in the environment. <strong>The</strong> organism is transmitted to<br />

PART 4 VACCINE-PREVENTABLE DISEASES 345<br />

4.15 Q FEVER


humans via the inhalation of infected aerosols or dust. Those most at risk include<br />

workers from the meat and livestock industries and shearers, with non-immune<br />

new employees or visitors being at highest risk of infection. Nevertheless,<br />

Q fever is not confined to occupationally exposed groups; there are numerous<br />

reports of sporadic cases or outbreaks in the general population in proximity to<br />

infected animals in stockyards, feedlots, processing plants or farms. Although<br />

most notifications occur among men from rural areas or with occupational<br />

exposure, a recent serosurvey from Queensland indicated a high rate of exposure<br />

among urban residents, including women and children. 25<br />

Use of Q fever vaccine in Australia can be considered in 4 periods: from 1991<br />

to 1993, when vaccine was used in a limited number of abattoirs; from 1994<br />

to 2000, when vaccination steadily increased to cover large abattoirs in most<br />

states; 26 from 2001 to 2006, during the period of the <strong>Australian</strong> Government<br />

sponsored National Q fever Management Program (NQFMP); 27 and the period<br />

since 2007 after the NQFMP finished, where the vaccination remains available<br />

on the private market. <strong>The</strong> NQFMP funded screening and vaccination of abattoir<br />

workers and shearers and then extended vaccination to farmers, their families<br />

and employees in the livestock-rearing industry. Following introduction of<br />

the program, the number of Q fever cases reported to the National Notifiable<br />

Diseases Surveillance System declined by over 50% (see Figure 4.15.1), 27 with<br />

the greatest reductions among young men aged 15–39 years, consistent with<br />

high documented vaccine uptake among abattoir workers. 26-28 As a result, other<br />

occupational groups as well as non-occupational animal exposures appear to be<br />

accounting for an increasing proportion of notifications. 8,29<br />

Figure 4.15.1: Q fever notifications for Australia, New South Wales and<br />

Queensland, 1991 to 2009 30<br />

Rate per 100 000 population<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

1991<br />

1992<br />

1993<br />

1994<br />

1995<br />

1996<br />

1997<br />

1998<br />

1999<br />

2000<br />

2001<br />

Year<br />

2002<br />

2003<br />

346 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

National Q Fever Management<br />

Program Phase 1<br />

Phase 2 implementation<br />

July 2002 to June 2005<br />

2004<br />

2005<br />

2006<br />

2007<br />

2008<br />

2009<br />

Qld<br />

NSW<br />

Aust


4.15.4 Vaccine<br />

• Q-Vax – CSL Limited (Q fever vaccine). Each 0.5 mL pre-filled<br />

syringe contains 25 µg purified killed suspension of Coxiella burnetii;<br />

thiomersal 0.01% w/v. Traces of formalin. May contain egg proteins.<br />

• Q-Vax Skin Test – CSL Limited (Q fever skin test). Each 0.5 mL liquid<br />

vial when diluted to 15 mL with sodium chloride contains 16.7 ng<br />

of purified killed suspension of C. burnetii in each diluted 0.1 mL<br />

dose; thiomersal 0.01% w/v before dilution. Traces of formalin. May<br />

contain egg proteins.<br />

<strong>The</strong> Q fever vaccine and skin test consist of a purified killed suspension of<br />

C. burnetii. It is prepared from the Phase I Henzerling strain of C. burnetii, grown<br />

in the yolk sacs of embryonated eggs. <strong>The</strong> organisms are extracted, inactivated<br />

with formalin, and freed from excess egg proteins by fractionation and<br />

ultracentrifugation. Thiomersal 0.01% w/v is added as a preservative.<br />

Phase I whole-cell vaccines have been shown to be highly antigenic and<br />

protective against challenge, both in laboratory animals and in volunteer trials. 31<br />

Serological response to the vaccine is chiefly IgM antibody to C. burnetii Phase I<br />

antigen. In subjects weakly seropositive before vaccination, the response is<br />

mainly IgG antibody to Phase I and Phase II antigens. 32 Lack of seroconversion<br />

is not a reliable marker of lack of vaccination. 31 Although the seroconversion<br />

rate may be low, long-term cell-mediated immunity develops33 and estimates of<br />

vaccine efficacy have ranged from 83 to 100%, based on the results of open and<br />

placebo-controlled trials, and post-marketing studies. 34-38 It is important that<br />

vaccination status is reported for all notified cases and apparent vaccine failures<br />

are investigated.<br />

It should be noted that vaccination during the incubation period of a natural<br />

attack of Q fever does not prevent the development of the disease. 31<br />

<strong>The</strong> Q fever vaccine and skin test are available for purchase in Australia through<br />

the private market. <strong>The</strong> <strong>Australian</strong> Government may fund the vaccine and skin<br />

test in emergency situations where there is a Q fever outbreak.<br />

4.15.5 Transport, storage and handling<br />

Transport the vaccine according to National vaccine storage guidelines: Strive for 5. 39<br />

Store at +2°C to +8°C. Do not freeze or store in direct contact with ice packs. If<br />

vaccine has been exposed to temperatures less than 0°C, do not use it. Protect<br />

from light.<br />

Diluted Q-Vax Skin Test should be freshly prepared, stored at +2°C to +8°C and<br />

used within 6 hours.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 347<br />

4.15 Q FEVER


4.15.6 Dosage and administration<br />

<strong>The</strong> dose of Q fever vaccine is 0.5 mL, to be given by SC injection, after<br />

ascertaining that serological and skin testing have been performed and that both<br />

tests are negative (see ‘Pre-vaccination testing’ below).<br />

Q fever vaccination and skin testing training is undertaken via an educational<br />

video available online. Please contact the manufacturer for access details.<br />

4.15.7 Recommendations<br />

Children aged


individuals, which can be accessed by registered users. If there is any doubt<br />

about serological or skin test results, testing should be repeated 2 to 3 weeks<br />

later (see below for interpretation).<br />

• Serological and skin test results should be taken into account, according to<br />

Table 4.15.1, before vaccination. Antibody studies were originally done by<br />

complement fixation (CF) tests at serum dilutions of 1 in 2.5, 5 and 10 against<br />

the Phase II antigen of C. burnetii. Although this is generally satisfactory,<br />

many testing laboratories now use enzyme immunoassay (EIA) or<br />

immunofluorescent antibody (IFA) to detect IgG antibody to C. burnetii as an<br />

indicator of past exposure. Subjects who are CF antibody positive at 1 in 2.5,<br />

IFA positive at 1 in 10 or more, or with a definite positive absorbance value in<br />

the EIA, should not be vaccinated.<br />

• Skin testing and interpretation should only be carried out by experienced<br />

personnel. Details of immunisation service providers trained in the<br />

administration of Q fever skin testing can be obtained online from the<br />

<strong>Australian</strong> Q Fever Register (www.qfever.org). Skin testing is performed<br />

by diluting 0.5 mL of the Q-Vax Skin Test in 14.5 mL of sodium chloride<br />

(injection grade). Diluted Q-Vax Skin Test should be freshly prepared, stored<br />

at +2°C to +8°C and used within 6 hours. A 0.1 mL dose of the diluted Q-Vax<br />

Skin Test is injected intradermally into the volar surface of the forearm.<br />

Commercial isopropyl alcohol skin wipes should not be used. If the skin is<br />

not visibly clean, then methylated spirits may be used. Skin reactions are<br />

common 3 to 4 days after skin testing; however, these reactions generally<br />

resolve by day 7 when the skin test is read. A positive reaction is indicated<br />

by any induration at the site of injection after 7 days. Individuals giving such<br />

a reaction must not be vaccinated, because they may develop severe local<br />

reactions.<br />

• <strong>The</strong> result of testing is considered ‘indeterminate’ when skin test induration<br />

is just palpable and the antibody test is either equivocal or negative, or when<br />

there is no skin induration and an equivocal antibody test (see Table 4.15.1).<br />

An indeterminate result, which occurs in only a small proportion of subjects,<br />

may be the consequence of past infection with Q fever. It may also merely<br />

indicate the presence in the subject of antibodies to antigens shared between<br />

C. burnetii and other bacteria. <strong>Australian</strong> Q fever vaccine providers have<br />

dealt with this finding in one of two ways:<br />

» Repeat the skin test and interpret as per the guidelines for initial testing.<br />

Collect serum 2 to 3 weeks later to look for a rise in titre of C. burnetii<br />

antibodies in the IFA test, using Phase I and Phase II antigens and<br />

immunoglobulin class analysis. A significant increase (defined as a<br />

4-fold rise in titre of paired sera) indicates previous Q fever infection and<br />

vaccination is then contraindicated.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 349<br />

4.15 Q FEVER


» Vaccinate the subject using SC injection of a 5 µg (0.1 mL) dose instead of<br />

a 25 µg (0.5 mL) dose of the vaccine. If there are no adverse events (e.g.<br />

severe local induration or severe systemic effects, perhaps accompanied<br />

by fever) 48 hours after the injection, a further 0.4 mL (20 µg) dose of<br />

the vaccine is given within the next 2 to 3 weeks, that is, before the<br />

development of cell-mediated immunity to the 1st dose.<br />

Table 4.15.1: Interpretation and action for serological and skin test results (with<br />

modifications from A guide to Q fever and Q fever vaccination<br />

(CSL Biotherapies, 2009) 6 )<br />

Serology Skin test Interpretation/Action<br />

Positive antibody test* Any skin test result Sensitised: do not vaccinate<br />

Equivocal antibody test †<br />

Negative antibody test #<br />

Positive ‡<br />

Borderline § or Negative <br />

Positive<br />

Borderline<br />

Negative<br />

350 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Sensitised: do not vaccinate<br />

Indeterminate (see above)<br />

Sensitised: do not vaccinate<br />

Indeterminate (see above)<br />

Non-immune: vaccinate<br />

* Positive antibody test: CF antibody or IFA positive (according to criteria used by diagnosing<br />

laboratory [see above]; or definite positive EIA absorbance value (according to manufacturer’s<br />

instructions)<br />

† Equivocal antibody test: CF antibody or IFA equivocal (according to criteria used by diagnosing<br />

laboratory); or equivocal EIA absorbance value (according to manufacturer’s instructions)<br />

‡ Positive skin test: induration present<br />

§ Borderline skin test: induration just palpable<br />

Negative skin test: no induration<br />

# Negative antibody test: CF antibody or IFA negative (according to criteria used by diagnosing<br />

laboratory); or definite negative EIA absorbance value (according to manufacturer’s instructions)<br />

Booster doses<br />

Immunity produced by the vaccine appears to be long-lasting (in excess of<br />

5 years). Until further information becomes available, revaccination or booster<br />

doses of the vaccine are not recommended because of the risk of accentuated<br />

local adverse events.<br />

4.15.8 Pregnancy and breastfeeding<br />

Q fever vaccine is not routinely recommended for pregnant or breastfeeding<br />

women.<br />

Q fever vaccine contains inactivated products; inactivated bacterial vaccines are<br />

not considered to be harmful in pregnancy. However, safety of the vaccine in<br />

pregnancy has not been established. No information is available on the use of<br />

Q fever vaccine during breastfeeding.


Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.15.9 Contraindications<br />

Q fever vaccine is contraindicated in the following groups:<br />

• persons with a history of laboratory-confirmed Q fever, or with medical<br />

documentation that supports a previous diagnosis of Q fever<br />

• persons shown to be immune by either serological testing or sensitivity to the<br />

organism by skin testing<br />

• persons who have been previously vaccinated against Q fever<br />

• persons with known hypersensitivity to egg proteins or any component of<br />

the vaccine (Q-Vax may contain traces of egg protein and formalin). 40<br />

<strong>The</strong>re is no information available on the accuracy of skin testing or the efficacy<br />

and safety of Q fever vaccine use in persons who are immunocompromised. In<br />

general, skin testing and Q fever vaccine should be avoided in such persons.<br />

<strong>The</strong>re are no data on the safety or efficacy of Q fever vaccine in children. Q fever<br />

vaccine is not recommended for use in those aged


skin test developed by the US National Institute of Health and the National<br />

Institute of Allergy and Infectious Diseases, 41 which was later combined with<br />

antibody testing in Australia, has largely eliminated reactions due to previous<br />

immune sensitisation. Despite this, the adverse experience from earlier American<br />

trials, 31 in which subjects were not pre-tested, were vaccinated repeatedly or were<br />

inoculated with vaccines of a different composition and larger bacterial mass,<br />

are still quoted in the general Q fever literature as representative of the broader<br />

experience with whole-cell Q fever vaccines.<br />

<strong>The</strong> second, much less frequent, pattern has been reported in people who are<br />

skin and antibody test-negative at the time of vaccination and who do not have<br />

any immediate reaction. Some 1 to 8 months after vaccination, some vaccine<br />

recipients, predominantly women, have developed an indurated lesion at the<br />

inoculation site. At the time when the indurated lesion develops, the original<br />

skin test site often becomes positive, presumably indicating a late developing<br />

cellular immune response. <strong>The</strong>se lesions are not fluctuant and do not progress<br />

to an abscess. Most gradually decline in size and resolve over some months<br />

without treatment. A few lesions have been biopsied or excised and have shown<br />

accumulations of macrophages and lymphocytes. 42,43<br />

4.15.12 Public health management of Q fever<br />

Q fever is a notifiable disease in all states and territories in Australia.<br />

Further instructions about the public health management of Q fever, including<br />

management of cases of Q fever, should be obtained from state/territory public<br />

health authorities (see Appendix 1 Contact details for <strong>Australian</strong>, state and territory<br />

government health authorities and communicable disease control).<br />

4.15.13 Variations from product information<br />

<strong>The</strong> product information for Q-Vax does not include the use of a reduced dose of<br />

vaccine in persons who have indeterminate results on either serological or skin<br />

testing. <strong>The</strong> ATAGI recommends instead that experienced Q fever vaccinators<br />

may elect to give reduced vaccine doses in subjects who have indeterminate<br />

results on either serological or skin testing.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

352 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.16 RABIES AND OTHER LYSSAVIRUSES (INCLUDING<br />

AUSTRALIAN BAT LYSSAVIRUS)<br />

4.16.1 Virology<br />

Lyssaviruses are single-stranded RNA viruses in the family Rhabdoviridae,<br />

genus Lyssavirus. <strong>The</strong>re are 12 known species within the genus Lyssavirus,<br />

including the classical rabies virus and other closely related lyssaviruses such as<br />

the <strong>Australian</strong> bat lyssavirus (ABLV) and European bat lyssaviruses. 1<br />

4.16.2 Clinical features<br />

Rabies is a zoonotic disease caused by human exposure to saliva or nerve<br />

tissue of an animal infected with rabies virus or other lyssaviruses. As the<br />

clinical disease caused by classical rabies virus and other lyssaviruses is<br />

indistinguishable, the term ‘rabies’ refers to disease caused by any of the known<br />

lyssavirus species. 2-5 Human exposure can occur via a scratch or bite that has<br />

broken the skin, or via direct contact with the mucosal surface of a person,<br />

such as nose, eye or mouth. Most human cases of rabies occur after animal<br />

bites – cases after animal scratches, the licking by animals of open wounds or<br />

contact of animal saliva with intact mucous membranes are very rare. Aerosol<br />

transmission has never been well documented in the natural environment. 6 <strong>The</strong>re<br />

has been transmission of rabies virus reported following tissue transplantation<br />

from donors who died with undiagnosed rabies. 7 Transmission of rabies virus to<br />

humans through unpasteurised milk may be possible; however, rare reports of<br />

transmission via this route have not been confirmed. 8<br />

Once a person is infected, the incubation period of rabies is usually 3 to 8 weeks,<br />

but can range from as short as a week to, on rare occasions, several years. <strong>The</strong><br />

risk of rabies is higher, and the incubation period shorter, after severe and<br />

multiple wounds proximate to the central nervous system (such as on the head<br />

and neck) and in richly innervated sites (such as the fingers).<br />

Rabies is almost invariably fatal. Typically, in the prodromal phase of rabies,<br />

which lasts up to 10 days, the patient may experience non-specific symptoms<br />

such as anorexia, cough, fever, headache, myalgia, nausea, sore throat, tiredness<br />

and vomiting. 9 Paraesthesiae and/or fasciculations at or near the site of the<br />

wound may be present at this stage. Anxiety, agitation and apprehension may<br />

also occur. Most rabies patients present with the furious or encephalitic form. 10<br />

In the encephalitic phase, objective signs of nervous system involvement include<br />

aerophobia, hydrophobia, bizarre behaviour, disorientation and hyperactivity.<br />

Signs of autonomic instability such as hypersalivation, hyperthermia and<br />

hyperventilation may occur. 10 <strong>The</strong> neurological status of the patient deteriorates<br />

over a period of up to 12 days, and the patient either dies abruptly from cardiac<br />

or respiratory arrest, or lapses into a coma.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 353<br />

4.16 RABIES AND OTHER<br />

LYSSAVIRUSES (INCLUDING<br />

AUSTRALIAN BAT LYSSAVIRUS)


4.16.3 Epidemiology<br />

<strong>The</strong> epidemiology of rabies varies depending on the lyssavirus species and the<br />

animal host. Lyssaviruses have been found in all continents, except Antarctica. 11<br />

Rabies that is due to the classical rabies virus and occurs in land dwelling<br />

(terrestrial) mammals is present throughout much of Africa, Asia, the Americas<br />

and Europe, where the virus is maintained in certain species of mammals,<br />

particularly dogs. In countries where rabies vaccination of domestic animals is<br />

widespread (North America and Europe), wild animals such as raccoons and<br />

foxes are important reservoirs. <strong>The</strong> continual maintenance of rabies in animal<br />

populations in these countries is referred to as enzootic rabies. Australia, New<br />

Zealand, Japan, Papua New Guinea and Pacific island nations are currently free<br />

of rabies in terrestrial mammals. However, a country’s status can change at any<br />

time. For example, in 2008 on the island of Bali, Indonesia, rabies was reported<br />

in dogs, with cases later reported in humans. 12 Prior to this, Bali had been<br />

considered free of rabies, although rabies was known to occur in other areas of<br />

Indonesia. 13<br />

In some parts of the world, bats are important reservoirs of classical rabies<br />

as well as other lyssaviruses, with bat lyssaviruses found in areas that are<br />

considered free from terrestrial rabies. ABLV was first reported in bats in 1996;<br />

since then, two cases of fatal encephalitis caused by ABLV have been reported<br />

in <strong>Australian</strong>s, one in 1996 and the other in 1998. 2,14 Both patients had been<br />

bitten by bats. Evidence of ABLV infection has since been identified in all four<br />

species of <strong>Australian</strong> fruit bats (flying foxes) and in several species of <strong>Australian</strong><br />

insectivorous bats. 4,15-17 It should therefore be assumed that all <strong>Australian</strong> bats<br />

have the potential to be infected with ABLV. Different regions in Australia have<br />

reported higher risk of potential ABLV exposures. 18,19 ABLV has not been isolated<br />

from bats outside Australia. However, closely related lyssaviruses are found in<br />

bats in other countries. For example, European bat lyssavirus 1 and European<br />

bat lyssavirus 2 have been isolated in bats in some parts of Europe. Four human<br />

deaths from European bat lyssavirus variants have been reported in Europe, all<br />

with no record of prophylactic rabies immunisation. 3,5 As such, bats anywhere in<br />

the world should be considered a potential source of lyssaviruses and a potential<br />

risk for acquiring rabies, depending on the exposure.<br />

Information on the global occurrence of rabies can be obtained from reputable<br />

international authorities. 11,20,21 In addition, advice on potential lyssavirus<br />

exposures and their management should be obtained by contacting the relevant<br />

<strong>Australian</strong> state/territory health authorities (see 4.16.12 Public health management<br />

of lyssavirus infections below).<br />

354 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.16.4 Rabies vaccines<br />

• Mérieux Inactivated Rabies Vaccine – Sanofi Pasteur Pty Ltd (human<br />

diploid cell vaccine [HDCV]). Lyophilised powder in a monodose<br />

vial with 1.0 mL distilled water as diluent. Each 1.0 mL reconstituted<br />

dose contains ≥2.5 IU inactivated rabies virus; 100–150 µg neomycin;<br />

≤70 mg human serum albumin; trace of phenol red (indicator). May<br />

contain trace amounts of bovine gelatin and β-propiolactone.<br />

• Rabipur Inactivated Rabies Virus Vaccine – CSL Limited/Novartis<br />

Vaccines and Diagnostics Pty Ltd (purified chick embryo cell<br />

vaccine [PCECV]). Lyophilised powder in a monodose vial with<br />

1.0 mL distilled water as diluent. Each 1.0 mL reconstituted dose<br />

contains ≥2.5 IU inactivated rabies virus; trace amounts of neomycin,<br />

chlortetracycline, trometamol, β-propiolactone, monopotassium<br />

glutamate and amphotericin B. May contain trace amounts of bovine<br />

gelatin and egg protein.<br />

<strong>The</strong>re are two inactivated rabies cell culture-derived vaccines available<br />

in Australia.<br />

<strong>The</strong> Mérieux vaccine is a lyophilised, stabilised suspension of inactivated Wistar<br />

rabies virus that has been cultured on human diploid cells and then inactivated<br />

by β-propiolactone. This human diploid cell vaccine (HDCV) is coloured offwhite,<br />

but after reconstitution with the diluent it turns a pinkish colour due to<br />

the presence of phenol red. <strong>The</strong> vaccine does not contain a preservative.<br />

Rabipur is a lyophilised, stabilised suspension of inactivated Flury LEP rabies<br />

virus that has been cultured on purified chick embryo cells and then inactivated<br />

by β-propiolactone. This purified chick embryo cell vaccine (PCECV) does not<br />

contain a preservative.<br />

Rabies vaccine is effective and safe when used for pre-exposure prophylaxis<br />

(PreP) and post-exposure prophylaxis (PEP) for rabies virus. 22,23 Although data on<br />

the effectiveness of rabies vaccine as prophylaxis against other lyssaviruses are<br />

limited, the available animal data and clinical experience support its use. 19,24-29<br />

4.16.5 Rabies immunoglobulin<br />

• Imogam Rabies Pasteurized – Sanofi Pasteur Pty Ltd (human rabies<br />

immunoglobulin [HRIG]). Each 1.0 mL contains IgG class human<br />

rabies antibodies with a minimum titre of 150 IU; 22.5 mg glycine;<br />

1 mg sodium chloride. Supplied in 2 mL vials.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 355<br />

4.16 RABIES AND OTHER<br />

LYSSAVIRUSES (INCLUDING<br />

AUSTRALIAN BAT LYSSAVIRUS)


Human rabies immunoglobulin (HRIG) is prepared by cold ethanol fractionation<br />

from the plasma of hyperimmunised human donors.<br />

4.16.6 Transport, storage and handling<br />

Rabies vaccine<br />

Transport according to National vaccine storage guidelines: Strive for 5. 30 Store at<br />

+2°C to +8°C. Do not freeze.<br />

Both rabies vaccines must be reconstituted by adding the entire contents of<br />

the diluent container to the vial and shaking until the powder is completely<br />

dissolved. Reconstituted vaccine should be used immediately.<br />

Human rabies immunoglobulin<br />

Transport according to National vaccine storage guidelines: Strive for 5. 30 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

HRIG should be used immediately once the vial is opened.<br />

4.16.7 Dosage and administration<br />

Rabies vaccine<br />

<strong>The</strong> dose of rabies vaccine for use in PreP and PEP is 1.0 mL, to be given by IM<br />

injection and is the same for infants, children and adults.<br />

HDCV can also be given by SC injection; however, if PCECV is inadvertently<br />

given via the SC route, the dose should be repeated.<br />

Note: Rabies vaccination administered via the intradermal (ID) route is not routinely<br />

used in Australia and is not recommended. <strong>The</strong> use of the ID route for rabies vaccination<br />

is the practitioner’s own responsibility, as rabies vaccines are not registered for use via<br />

this route in Australia. ID administration is particularly not recommended for postexposure<br />

prophylaxis. For detailed information on the restrictions that apply to the use<br />

of ID vaccination, if undertaken for PreP, see ‘Pre-exposure prophylaxis administered via<br />

the intradermal route’ below.<br />

Rabies vaccine should be given in the deltoid area, as rabies virus neutralising<br />

antibody (VNAb) titres may be reduced after administration in other sites.<br />

In particular, vaccine should never be given in the buttock, as failure of preexposure<br />

prophylaxis has been reported when given by this route. 31 In infants<br />


of an even more accelerated schedule for those with limited time before travel to<br />

a rabies-enzootic area.<br />

Post-exposure prophylaxis (PEP)<br />

In persons previously unvaccinated, the recommended schedule for postexposure<br />

prophylaxis (PEP) for immunocompetent persons consists of 4 doses<br />

of vaccine. <strong>The</strong> 1st dose of vaccine is given as soon as practicable (day 0), and<br />

subsequent doses are given on days 3, 7 and 14; deviations of a few days from<br />

this schedule are probably unimportant. 22<br />

<strong>The</strong> recommended schedule for PEP for previously unvaccinated<br />

immunocompromised persons consists of 5 doses of vaccine. <strong>The</strong> 1st dose of<br />

vaccine is given as soon as practicable (day 0), and subsequent doses are given<br />

on days 3, 7, 14 and 28; deviations of a few days from this schedule are probably<br />

unimportant.<br />

<strong>The</strong> recommended schedule for PEP for people who have been previously<br />

vaccinated against rabies consists of 2 doses of rabies vaccine on days 0 and 3<br />

(noting caveats in Figures 4.16.1 and 4.16.2).<br />

For more detailed information see 4.16.8 Recommendations below.<br />

Human rabies immunoglobulin<br />

When HRIG is indicated, the dose is 20 IU per kilogram of body mass and is the<br />

same for infants, children and adults. HRIG should be administered at the same<br />

time as the 1st dose (day 0) of rabies vaccine. Do not administer HRIG if 8 days<br />

or more have elapsed since the 1st dose of vaccine, as the HRIG may interfere<br />

with the immune response to the vaccine. For more detailed information see<br />

4.16.8 Recommendations below.<br />

HRIG should be infiltrated in and around all wounds using as much of the calculated<br />

dose as possible, and the remainder of HRIG administered intramuscularly at a<br />

site away from the rabies vaccine injection site. If the wounds are severe and the<br />

calculated volume of HRIG is inadequate for complete infiltration of all wounds<br />

(e.g. extensive dog bites in a young child), the HRIG should be diluted in saline<br />

to make up an adequate volume for the careful infiltration of all wounds.<br />

Wounds to fingers and hands may be small, particularly following exposures to<br />

bats, and infiltration of HRIG into these wounds is likely to be both technically<br />

difficult and painful for the recipient. 32 However, due to the extensive nerve<br />

supply to these sites9,10,33 it is important that as much of the calculated dose of<br />

HRIG as possible should be infiltrated into finger and hand wounds using either<br />

a 25 or 26 gauge needle. To avoid the development of a compartment syndrome,<br />

the HRIG should be infiltrated very gently, and should not cause the adjacent<br />

finger tissue to go frankly pale or white. It may be necessary to give a ring-block<br />

using a local anaesthetic. 32<br />

PART 4 VACCINE-PREVENTABLE DISEASES 357<br />

4.16 RABIES AND OTHER<br />

LYSSAVIRUSES (INCLUDING<br />

AUSTRALIAN BAT LYSSAVIRUS)


Interchangeability of rabies vaccines<br />

<strong>The</strong> World Health Organization (WHO) does not recommend interchanging<br />

rabies cell culture-derived vaccines (CCV), but states that, in situations where<br />

it is unavoidable, a PreP or PEP course can be completed with an alternative<br />

rabies CCV, providing the vaccine is WHO-endorsed (also termed ‘prequalified’).<br />

22 Various international vaccine advisory groups state that rabies<br />

CCV are interchangeable. This is supported by the similarities in tissue culture<br />

vaccine production methods as well as antibody responses and adverse<br />

reactions following vaccination. In one study that specifically assessed the<br />

interchangeability of HDCV and PCECV, 165 subjects were randomised to receive<br />

rabies PreP (days 0, 7 and 21–28) using either HDCV or PCECV. 34 One year<br />

following PreP, each group received 1 or 2 booster doses of PCECV. <strong>The</strong> booster<br />

dose resulted in an anamnestic response (geometric mean titre several orders of<br />

magnitude >0.5 IU/mL) in all subjects by day 7, independent of the vaccine that<br />

was used to deliver the primary course. It is expected that this response would be<br />

similar with other rabies CCV.<br />

4.16.8 Recommendations<br />

Measures to avoid potential rabies virus and other lyssavirus (including ABLV)<br />

exposures<br />

Travellers to rabies-enzootic regions should be advised to avoid close contact<br />

with either wild or domestic animals; this is particularly important for<br />

children. 35-37 <strong>The</strong>y should be advised about pre-travel (i.e. pre-exposure) rabies<br />

vaccination (or, if appropriate, booster doses), and on what to do should they<br />

be either bitten or scratched by an animal while abroad. 37-41 It is recommended<br />

that prior to travel, travellers be educated regarding first aid treatment for rabies<br />

exposures, irrespective of prior vaccination.<br />

Recommendations to decrease the risk of exposure to rabies include:<br />

• Do not allow young children to feed, pat or play with animals. <strong>The</strong> height of<br />

young children makes bites to the face and head more likely.<br />

• Avoid contact with stray dogs or cats. Remain vigilant when walking,<br />

running or cycling.<br />

• Do not carry food, and do not feed or pat monkeys, even in popular areas<br />

around temples or markets where travellers may be encouraged to interact<br />

with the monkeys. In particular, avoid focusing attention on monkeys<br />

carrying their young, as they may feel threatened and bite suddenly.<br />

In addition, contact with bats should be avoided anywhere in the world,<br />

including Australia. Only appropriately vaccinated and trained persons should<br />

handle bats. If bats must be handled, safety precautions, such as wearing<br />

protective gloves and clothing, should be observed.<br />

358 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Pre-exposure prophylaxis for rabies virus and other lyssaviruses (including ABLV)<br />

PreP with rabies vaccine is recommended for:<br />

• persons liable to receive bites or scratches from bats (this includes bat<br />

handlers, veterinarians, wildlife officers and others who come into direct<br />

contact with bats) in any country, including Australia<br />

• travellers and expatriates who will be spending time in rabies-enzootic areas;<br />

PreP should occur following a risk assessment that takes into consideration<br />

the likelihood of interaction with animals and access to emergency medical<br />

attention<br />

• persons working with terrestrial animals in rabies-enzootic areas<br />

• research laboratory personnel working with any live lyssaviruses.<br />

Parents travelling with children to rabies-enzootic areas should consider PreP for<br />

younger children, as many children, if bitten by dogs, are often bitten on the face<br />

and hands because they are at an optimal height for such contact.<br />

Serological testing to confirm seroconversion is only necessary in certain<br />

circumstances (see ‘Serological testing following rabies vaccination’ below). 22<br />

PreP simplifies the management of a subsequent exposure because fewer doses<br />

of vaccine are needed and because rabies immunoglobulin (RIG) is not required<br />

(see ‘Post-exposure prophylaxis for rabies virus and other lyssavirus (including<br />

ABLV) exposures’ below). This is particularly important as RIG (human or<br />

equine) is often difficult to obtain in many developing countries and its safety<br />

may not be guaranteed.<br />

Pre-exposure prophylaxis administered via the intradermal route<br />

Intradermal PreP is not recommended because, although initial antibody titres<br />

may be higher, titres at 14 days are lower and wane more rapidly after ID<br />

administration of rabies vaccine than after either IM or SC administration. <strong>The</strong>re<br />

may also be a slow initial immune response following exposure to rabies virus<br />

in those given ID rabies vaccine. 42-44 For these reasons, it is strongly recommended<br />

that the IM route (IM or SC if HDCV is used) be used for pre-exposure prophylaxis. (See<br />

also 4.16.7 Dosage and administration above.)<br />

However, if ID rabies PreP is considered (using a dose of 0.1 mL on days 0, 7 and<br />

28) it is essential that:<br />

• it is given by vaccine providers with not only expertise in, but also regular<br />

practice of, the ID technique<br />

• it is not administered to anyone who is immunocompromised<br />

• it is not administered to persons taking either chloroquine or other<br />

antimalarials structurally related to chloroquine (e.g. mefloquine), at either<br />

the time of, or within a month following, vaccination45 • any remaining vaccine is discarded at the end of the session during which the<br />

vial is opened (8 hours)<br />

PART 4 VACCINE-PREVENTABLE DISEASES 359<br />

4.16 RABIES AND OTHER<br />

LYSSAVIRUSES (INCLUDING<br />

AUSTRALIAN BAT LYSSAVIRUS)


• the rabies VNAb level is checked 14 to 21 days following completion of the<br />

pre-exposure course of ID vaccine (see ‘Serological testing following rabies<br />

vaccination’ below)<br />

• it is only used for PreP for classical rabies exposures (there are no data on the<br />

protection provided by ID rabies vaccination for the prevention of infection<br />

with other lyssaviruses, including ABLV).<br />

Post-exposure prophylaxis for rabies virus and other lyssavirus (including ABLV)<br />

exposures<br />

PEP for rabies virus and other lyssavirus exposures consists of prompt wound<br />

management, vaccine and HRIG administration. <strong>The</strong> appropriate combination<br />

of these components depends on the extent of the exposure, the animal source<br />

of the exposure, the person’s immune status and their previous vaccination<br />

history. <strong>The</strong> different PEP pathways are described in more detail below and PEP<br />

management algorithms are outlined in Figures 4.16.1 and 4.16.2.<br />

Types of potential rabies virus and other lyssavirus (including ABLV) exposures<br />

Three different categories of lyssavirus exposure are outlined in Table 4.16.1,<br />

based on those already described by the WHO. 22 <strong>The</strong> appropriate PEP pathway<br />

following each of the different exposure categories varies depending on<br />

whether the source of exposure was a terrestrial animal or a bat. Different<br />

PEP management pathways following potential bat exposures compared with<br />

terrestrial animal exposures are required because the risk from wounds from<br />

bats is often difficult to determine due to the limited injury inflicted and there<br />

is evidence that superficial bat exposures are more likely to result in human<br />

infection. 31<br />

An algorithm detailing the appropriate PEP pathway following potential classical<br />

rabies virus exposure from a terrestrial animal is provided in Figure 4.16.1; an<br />

algorithm for use following potential lyssavirus exposure from a bat is provided<br />

in Figure 4.16.2.<br />

Table 4.16.1: Lyssavirus exposure categories, to be used in conjunction with<br />

Figure 4.16.1 or 4.16.2 to determine appropriate post-exposure<br />

prophylaxis<br />

Type of exposure Description<br />

Category I Touching or feeding animals, licks on intact skin, as well<br />

as exposure to blood, urine or faeces or to an animal that<br />

has been dead for more than 4 hours<br />

Category II Nibbling of uncovered skin, minor scratches or abrasions<br />

without bleeding<br />

Category III Single or multiple transdermal bites or scratches,<br />

contamination of mucous membrane with saliva from<br />

licks, licks on broken skin<br />

Source: Modified from WHO 2010 22<br />

360 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


<strong>The</strong> relevant state/territory health authority should be contacted about any<br />

potential exposure sustained from a terrestrial animal in a rabies-enzootic area, or<br />

any potential exposure sustained from a bat anywhere in the world46 (see 4.16.12<br />

Public health management of lyssavirus infections below). Dogs and monkeys are the<br />

usual exposures in Asia, Africa and Central and South America, but exposures to<br />

other terrestrial mammals and bats must also be assessed for potential classical<br />

rabies virus transmission. If a traveller presents >10 days after being bitten or<br />

scratched by either a domestic dog, cat or ferret in a rabies-enzootic country, and<br />

it can be reliably ascertained that the animal has remained healthy (>10 days after<br />

the exposure), PEP is not required. Otherwise, PEP appropriate for the category<br />

and source of exposure (see Figure 4.16.1 or 4.16.2) should be administered, even<br />

if there has been a considerable delay in reporting the incident.<br />

<strong>The</strong> relevant state/territory veterinary or health authority should be contacted<br />

regarding any potential exposure to <strong>Australian</strong> bats (for ABLV) (see 4.16.12 Public<br />

health management of lyssavirus infections below). This includes situations where<br />

the category of the exposure is unsure, for example, for a person or child who<br />

has woken up with a bat present in a confined space but with no recollection of<br />

contact. If possible, and without placing others at risk of exposure, the bat should<br />

be kept and arrangements promptly made for testing the bat for ABLV. Following<br />

wound management (see below), the administration of HRIG and rabies vaccine<br />

can be withheld if the bat’s ABLV status will be available within 48 hours of<br />

the exposure. If the result will not be available within 48 hours, the appropriate<br />

post-exposure prophylaxis should begin as soon as is practicable, following the<br />

bat exposure algorithm as outlined in Figure 4.16.2. Where a bat is tested at a<br />

reference laboratory and later found to be negative for ABLV, then PEP for the<br />

person exposed to that bat can be discontinued.<br />

Wound management in post-exposure prophylaxis<br />

One of the most vital steps following a potential rabies virus or other lyssavirus<br />

exposure is wound management. Immediate and thorough washing of all bite<br />

wounds and scratches with soap and water, and the application of a virucidal<br />

preparation such as povidone-iodine solution after the washing, are important<br />

measures in the prevention of rabies. Consideration should also be given to the<br />

possibility of tetanus and other wound infections, and appropriate measures<br />

taken. Primary suture of a bite from a potentially rabid animal should be<br />

avoided. Bites should be cleaned, debrided and infiltrated well with HRIG, when<br />

indicated (see Figure 4.16.1 or 4.16.2).<br />

Post-exposure prophylaxis of persons who are previously unvaccinated<br />

Vaccine<br />

After performing wound management, rabies vaccine should be administered<br />

with or without HRIG (see ‘Human rabies immunoglobulin’ below), depending<br />

on the category and source of exposure, as outlined in Figure 4.16.1 or Figure<br />

4.16.2, and described below.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 361<br />

4.16 RABIES AND OTHER<br />

LYSSAVIRUSES (INCLUDING<br />

AUSTRALIAN BAT LYSSAVIRUS)


Persons who have not previously received a complete rabies vaccine course, and<br />

are immunocompetent, should receive a total of 4 doses of rabies vaccine (see<br />

4.16.7 Dosage and administration above). Although no clinical trial has assessed the<br />

efficacy of rabies vaccine, the rationale supporting the use of a 4-dose schedule<br />

in immunocompetent persons is based on 11 studies where the immunogenicity<br />

of either cell culture-derived vaccine was consistently >0.5 IU/mL by day 30<br />

(after 4 doses) and, in a majority of participants, was >0.5 IU/mL by day 14<br />

(after 3 doses). Antibody responses observed after the 4th and 5th doses were<br />

both several orders of magnitude larger than the WHO cut-off of 0.5 IU/mL and<br />

were similar in value. As the additional immune boosting following a 5th dose is<br />

minimal, a 5th dose is not required in immunocompetent persons. 47-57<br />

Persons who have not previously received a complete rabies vaccine<br />

course and who have either an immunocompromising illness, or are taking<br />

immunosuppressant medications, should receive a 5-dose vaccine schedule<br />

(see 4.16.7 Dosage and administration above). 58-60 <strong>The</strong> rabies VNAb titre should be<br />

measured 14 to 21 days after the 5th dose and a further dose given if the titre is<br />

reported as inadequate (i.e.


Although data are limited on the effectiveness of rabies vaccine and HRIG as PEP<br />

against infection with lyssaviruses other than classical rabies virus, the available<br />

animal data and clinical experience support their use. 19,24-29<br />

Post-exposure prophylaxis of persons who have been previously vaccinated<br />

Wound management must still be carried out irrespective of prior rabies<br />

vaccination.<br />

Persons who have evidence of a previous completed recommended PreP or PEP<br />

regimen, or who have a previously documented adequate VNAb titre, require a<br />

total of 2 doses of rabies vaccine (see Figure 4.16.1 or Figure 4.16.2). This includes<br />

immunocompromised individuals; however, VNAb levels should be checked<br />

after the 2nd dose to ensure they are adequate (see ‘Serological testing following<br />

rabies vaccination’ below).<br />

Note: PreP or PEP vaccine administered via the ID route is not considered<br />

appropriate previous vaccination unless documentation of an adequate VNAb<br />

titre is available (see ‘Serological testing following rabies vaccination’ below).<br />

HRIG is not required and should not be administered, as its use may suppress<br />

the level of anamnestic response and circulating VNAb.<br />

In cases where a person’s vaccination status is uncertain because the<br />

documentation of a full course of rabies vaccine is not available, the full PEP<br />

regimen should be administered.<br />

Post-exposure prophylaxis commenced overseas<br />

<strong>Australian</strong>s travelling overseas who are exposed to a potentially rabid animal<br />

(including bats from any country) may be given PEP using vaccines and<br />

schedules not used in Australia. In very rare circumstances, if an older nerve<br />

tissue-derived rabies vaccine has been administered, any doses given should be<br />

disregarded (see Table 4.16.2). However, it is most likely that a person vaccinated<br />

overseas will have received a cell culture-derived vaccine (see ‘Interchangeability<br />

of rabies vaccines’ in 4.16.7 Dosage and administration above). 22,34 If a person<br />

has received a cell culture-derived vaccine abroad, it is recommended that the<br />

standard post-exposure prophylaxis regimen be continued in Australia with<br />

either HDCV or PCECV.<br />

WHO-approved post-exposure rabies vaccination regimens include:<br />

• Zagreb (2 doses on day 0, doses on days 7 and 21: annotated as 2-0-1-1)<br />

• Essen (doses given on days 0, 3, 7, 14 and 28 (or 30): annotated as 1-1-1-1-1)<br />

• Modified Essen (doses given on days 0, 3, 7 and 14: annotated as 1-1-1-1).<br />

If the PEP was started overseas but HRIG or equine RIG was not given, and the<br />

person presents in Australia within 7 days of commencing PEP, HRIG should be<br />

given as soon as is practicable (and within 7 days of the 1st rabies vaccine dose).<br />

If the person presents in Australia 8 days or more after commencing PEP, then<br />

HRIG should not be administered and the appropriate number of remaining<br />

doses of rabies vaccine administered.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 363<br />

4.16 RABIES AND OTHER<br />

LYSSAVIRUSES (INCLUDING<br />

AUSTRALIAN BAT LYSSAVIRUS)


For these and other scenarios that may arise, Table 4.16.2 outlines the most<br />

common PEP regimens that may be commenced overseas and the recommended<br />

schedule to complete PEP in Australia.<br />

In the case of PEP commenced overseas, every traveller should be advised to<br />

request a PEP certificate from the vaccination centre and to obtain or record the<br />

following information (preferably in English):<br />

• the contact details for the clinic attended (telephone and email address)<br />

• the batch and source of RIG used (Note: equine RIG rather than human RIG<br />

may be used in some countries)<br />

• the volume of RIG administered<br />

• the type of cell culture vaccine used<br />

• the vaccine batch number<br />

• the number of vials used<br />

• the route of vaccine administration<br />

• the date of RIG and/or vaccine administration.<br />

<strong>The</strong>se details help inform decisions about PEP on return home.<br />

364 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Table 4.16.2: Post-exposure prophylaxis commenced overseas and<br />

recommended completion in Australia<br />

Vaccine type/route<br />

administered overseas/<br />

rabies immunoglobulin<br />

(RIG)<br />

Rabies vaccine schedule<br />

in Australia<br />

Nerve tissue vaccine Recommence schedule<br />

starting from day 0<br />

Unsure/unknown/poor<br />

documentation<br />

Well documented, RIG<br />

(equine or human) given,<br />

plus vaccine given either<br />

IM or ID<br />

2 vaccine doses given IM<br />

on day 0<br />

Irrespective of whether RIG<br />

(equine or human) was<br />

administered at same time<br />

as the 1st doses of vaccine<br />

Immunocompromised<br />

with vaccines<br />

administered ID<br />

Recommence schedule<br />

starting from day 0<br />

Align with nearest due<br />

dose and resume schedule<br />

administering vaccine IM<br />

(IM or SC if HDCV used)<br />

Give a further 2 doses; the<br />

1st dose on day 7 and the<br />

2nd dose on day 14<br />

Irrespective of number<br />

of previous doses,<br />

administer a 5-dose<br />

schedule IM (IM or SC if<br />

HDCV used) and check<br />

serology (see ‘Serological<br />

testing following rabies<br />

vaccination’ below)<br />

HRIG<br />

Category III terrestrial<br />

animal exposures and<br />

Category II and III bat<br />

exposures only*<br />

Administer HRIG if no<br />

RIG already given<br />

Do not give HRIG if 8 days<br />

or more since 1st dose of<br />

vaccine (day 0)<br />

Administer HRIG if no<br />

RIG already given<br />

Do not give HRIG if 8 days<br />

or more since 1st dose of<br />

vaccine (day 0)<br />

No HRIG needed<br />

Administer HRIG if no<br />

RIG already given<br />

Do not give HRIG if 8 days<br />

or more since 1st doses of<br />

vaccine (day 0)<br />

Administer HRIG if no<br />

RIG already given<br />

Do not give HRIG if 8 days<br />

or more since 1st dose of<br />

vaccine (day 0)<br />

* See Table 4.16.1 Lyssavirus exposure categories and Figures 4.16.1 and 4.16.2 for further information<br />

of PEP pathways, including HRIG administration following either a terrestrial animal or bat<br />

exposure.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 365<br />

4.16 RABIES AND OTHER<br />

LYSSAVIRUSES (INCLUDING<br />

AUSTRALIAN BAT LYSSAVIRUS)


Figure 4.16.1: Post-exposure prophylaxis algorithm for potential exposure to<br />

classical rabies virus from a terrestrial animal overseas<br />

Potential exposure from a terrestrial animal in a rabies-enzootic area<br />

Category I<br />

• Touching or feeding animals,<br />

licks on intact skin<br />

• Exposure to blood, urine or<br />

faeces or to an animal that<br />

has been dead f .<br />

No prophylaxis is<br />

required if contact<br />

history is reliable<br />

Vaccinate §<br />

4 doses administered IM on<br />

days 0, 3, 7 and 14. Human<br />

rabies immunoglobulin (HRIG)<br />

is not indicated.<br />

Category II<br />

Nibbling of uncovered skin,<br />

minor scratches or abrasions<br />

without bleeding<br />

Non-immune, ‡<br />

immunocompetent<br />

If further exposures in the future<br />

Treat as previously immunised and<br />

follow algorithm as above<br />

If ongoing occupational exposure<br />

risk – see Booster algorithm<br />

* If in doubt, treat as non-immune.<br />

Previously<br />

immunised* †<br />

Vaccinate <br />

Both immunocompetent and<br />

immunocompromised persons – 2 doses<br />

delivered IM on days 0 and 3. HRIG is<br />

not indicated.<br />

366 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Category III<br />

• Single or multiple<br />

transdermal bites or scratches<br />

• Contamination of mucous<br />

membrane with saliva<br />

from licks<br />

• Licks on broken skin<br />

Non-immune , ‡<br />

immunocompetent<br />

§<br />

Vaccinate and administer HRIG<br />

HRIG is administered only once, and as<br />

soon as possible after the initiation of<br />

PEP (HRIG is not indicated beyond the<br />

7th day after the 1st vaccine dose on<br />

day 0). Rabies vaccination is 4 doses<br />

administered IM on days 0, 3, 7 and 14.<br />

† Previously immunised – documentation of a completed recommended PreP or PEP rabies<br />

vaccine regimen. This is irrespective of the time period since the last dose was administered. This<br />

may either be a completed primary pre-exposure course or post-exposure course and includes<br />

those where subsequent boosting has occurred, or documented rabies antibody (VNAb) titres of<br />

≥0.5 IU/mL.<br />

‡ Non-immune – person who has never received pre- or post-exposure immunisation with rabies<br />

vaccine, has had incomplete/inadequate primary vaccination course.<br />

§ Immunocompromised persons, not previously vaccinated, should receive 5 doses of vaccine<br />

on days 0, 3, 7, 14 and 28. Serology should be checked 14 to 21 days post dose 5 and a further<br />

dose offered if the result is


Figure 4.16.2: Post-exposure prophylaxis algorithm for potential exposure to<br />

lyssaviruses from bats in Australia or overseas<br />

Category I<br />

• Touching or feeding<br />

animals, licks on<br />

intact skin<br />

• Exposed to blood,<br />

urine or faeces or to<br />

an animal that has been<br />

dead for >4 hours<br />

No prophylaxis is<br />

required if contact<br />

history is reliable.<br />

Potential exposure from a bat (Australia or overseas)<br />

Category II or III<br />

Nibbling of uncovered skin, any scratches or abrasions with/without<br />

bleeding, single or multiple transdermal bites or scratches, contamination<br />

of mucous membrane with saliva from licks, or licks on broken skin<br />

* †<br />

Previously immunised<br />

Vaccinate <br />

Both immunocompetent<br />

and immunocompromised<br />

persons – 2 doses delivered<br />

IM on days 0 and 3. Human<br />

rabies immunoglobulin<br />

(HRIG) is not indicated.<br />

Perform serology<br />

i) Every 6 months for laboratory staff at risk<br />

ii) Every 2 years for veterinary workers, bat handlers or<br />

any other workers who are likely to need to handle bats.<br />

Give a single booster dose<br />

If further exposure give PEP<br />

as above<br />

Ongoing occupational exposure risk<br />

L<br />

Non-immune, ‡<br />

immunocompetent<br />

§ §<br />

Vaccinate and administer HRIG<br />

HRIG is administered only once, and as<br />

soon as possible after the initiation of<br />

PEP (HRIG is not indicated beyond the<br />

7th day after the 1st vaccine dose on<br />

day 0). Rabies vaccination is 4 doses<br />

administered IM on days 0, 3, 7 and 14.<br />

No further action until either<br />

• exposure, then give PEP as above<br />

OR<br />

• time period elapses as above for<br />

serology – undertake VNAb serology<br />

* If in doubt, treat as non-immune.<br />

† Previously immunised – documentation of a completed recommended PreP or PEP rabies vaccine<br />

regimen. This is irrespective of the time period since the last dose was administered. This may either<br />

be a completed primary pre-exposure course or post-exposure course and includes those where<br />

subsequent boosting has occurred, or documented rabies antibody (VNAb) titres of ≥0.5 IU/mL.<br />

‡ Non-immune – person who has never received pre- or post-exposure immunisation with rabies<br />

vaccine or has had incomplete/inadequate primary vaccination course.<br />

§ Immunocompromised persons, not previously vaccinated, should receive 5 doses of vaccine on days<br />

0, 3, 7, 14 and 28. Serology should be checked 14 to 21 days post dose 5 and a further dose offered<br />

if the result is


Booster doses<br />

A recent WHO position paper applied a quality assessment of a moderate level<br />

of scientific evidence to support that cell culture-derived rabies vaccines induce<br />

long-term immunity of at least 10 years. 22<br />

<strong>The</strong> WHO states that booster doses are not required for persons who are<br />

travelling to, or living in, an area of high rabies risk and who have completed<br />

a primary course, either pre- or post-exposure, using either of the currently<br />

available cell culture-derived vaccines. 22<br />

Booster doses of rabies vaccine are recommended for immunised persons who<br />

have ongoing occupational exposure to lyssaviruses in Australia or overseas62 (see Figure 4.16.3).<br />

<strong>The</strong>se include:<br />

• Persons who work with live lyssaviruses in research laboratories who should<br />

have rabies neutralising antibody titres measured every 6 months. If the titre<br />

is reported as inadequate (


Figure 4.16.3: Booster algorithm for persons at ongoing risk of exposure to<br />

either rabies or other lyssaviruses, including <strong>Australian</strong> bat<br />

lyssavirus (ABLV)<br />

Rabies or bat lyssavirus (including ABLV) booster algorithm<br />

Ongoing occupational exposure risk<br />

Perform serology<br />

i)<br />

ii) Every 2 years for veterinary workers, bat handlers or any other workers<br />

who are likely to need to handle bats.<br />

Viral neutralising antibodies<br />

(<br />

Give a single booster dose*<br />

If further exposure, give PEP as<br />

per rabies or bat lyssavirus postexposure<br />

algorithms<br />

No further action until either<br />

• further exposure, then give<br />

PEP as per rabies or bat<br />

lyssavirus post-exposure<br />

algorithms<br />

OR<br />

• time period elapses as above<br />

for serology – undertake<br />

VNAb serology<br />

* Immunocompromised patients’ serology should be checked 14 to 21 days post booster dose and a<br />

further dose offered if the result remains


to 2 years, depending on the risk of exposure, to assess the need for booster<br />

vaccination (see ‘Booster doses’ above).<br />

4.16.9 Pregnancy and breastfeeding<br />

Rabies vaccine and HRIG are recommended in pregnant or breastfeeding women<br />

following a potential exposure to rabies virus, ABLV or another bat lyssavirus64,65 (see 4.16.8 Recommendations above and 3.3 Groups with special vaccination<br />

requirements, Table 3.3.1 Recommendations for vaccination in pregnancy).<br />

4.16.10 Contraindications<br />

<strong>The</strong>re are no absolute contraindications to use of either rabies vaccine or HRIG as<br />

post-exposure prophylaxis in persons with a potential exposure to rabies or other<br />

lyssaviruses, including ABLV. This is because rabies disease is almost always<br />

lethal.<br />

Persons with an anaphylactic sensitivity to eggs, or to egg proteins, should not<br />

receive PCECV. HDCV should be used instead.<br />

See also 4.16.11 Adverse events below.<br />

4.16.11 Adverse events<br />

Cell culture-derived vaccines are generally well tolerated. In a large study, the<br />

following adverse events were reported after administration of HDCV to adults:<br />

sore arm (in 15 to 25% of vaccine recipients), headache (in 5 to 8%), malaise,<br />

nausea or both (in 2 to 5%), and allergic oedema (in 0.1%). 66 Similar adverse event<br />

profiles have been reported for the PCECV; these reactions occur at the same<br />

rates in children. 22,23,67-72<br />

Although anaphylactic reactions are rare (approximately 1 per 10 000<br />

vaccinations) following administration of HDCV, approximately 6% of persons<br />

receiving booster doses may experience allergic reactions. <strong>The</strong> reactions typically<br />

occur 2 to 21 days after a booster dose, and are characterised by generalised<br />

urticaria, sometimes with arthralgia, arthritis, oedema, nausea, vomiting, fever<br />

and malaise. 66 <strong>The</strong>se reactions are not life-threatening; they have been attributed<br />

to the presence of β-propiolactone-altered human albumin in the implicated<br />

vaccines. 66<br />

HRIG has an excellent safety profile and, in general, no chance of immediate<br />

hypersensitivity reactions as is more often the case with some equine sources of<br />

rabies immunoglobulin. 22<br />

Management of adverse events<br />

Once initiated, rabies post-exposure prophylaxis should not be interrupted or<br />

discontinued because of local reactions or mild systemic reactions. Such reactions<br />

can usually be managed with simple analgesics.<br />

Because rabies disease is almost always lethal, the recommended vaccination<br />

regimens, in particular the PEP regimen, should be continued even if a significant<br />

370 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


allergic reaction occurs following a dose of rabies vaccine. Antihistamines can be<br />

administered in an attempt to ameliorate any subsequent reactions.<br />

A patient’s risk of developing either lyssavirus infection or rabies must be<br />

carefully considered before deciding to discontinue vaccination.<br />

4.16.12 Public health management of lyssavirus infections<br />

Classical rabies virus and ABLV virus infections in humans are notifiable diseases<br />

in all states and territories in Australia.<br />

Other lyssavirus cases that do not meet the case definition for ABLV or rabies<br />

virus infection are also notifiable in all states and territories in Australia.<br />

Detailed information regarding the management of disease from rabies and other<br />

lyssaviruses, including ABLV, can be found in the national guidelines for public<br />

health units46 (www.health.gov.au/cdnasongs).<br />

Both HRIG and rabies vaccine are available for PEP from the relevant state/<br />

territory health authorities (see Appendix 1 Contact details for <strong>Australian</strong>, state and<br />

territory government health authorities and communicable disease control).<br />

4.16.13 Variations from product information<br />

Neither of the product information sheets for the two vaccines available in<br />

Australia mentions that they can be used for both PreP and PEP for ABLV<br />

exposures. <strong>The</strong> ATAGI instead recommends that, where indicated, either<br />

of the available rabies vaccines can be used as PreP or PEP as per 4.16.8<br />

Recommendations above.<br />

<strong>The</strong> product information for HDCV recommends a routine 6th dose at 90 days<br />

in a PEP regimen. <strong>The</strong> ATAGI recommends instead that a 4-dose schedule be<br />

used for PEP in immunocompetent persons. Further doses should be offered to a<br />

person who is immunocompromised and who has an inadequate antibody level<br />

following the 5-dose PEP regimen.<br />

<strong>The</strong> product information for HDCV also recommends a pre-exposure booster<br />

after a year. <strong>The</strong> ATAGI instead recommends boosters between 6 months and<br />

2 years for persons at continuing occupational risk (see ‘Booster doses’ in 4.16.8<br />

Recommendations above).<br />

<strong>The</strong> product information for PCECV recommends a routine 5th dose at<br />

28 days in a PEP regimen and the product information for HDCV recommends<br />

a routine 5th and 6th dose at days 30 and 90, respectively, in a PEP regimen.<br />

<strong>The</strong> ATAGI recommends instead that a 4-dose schedule with either cell culturederived<br />

vaccine be used for PEP in immunocompetent persons. A 5th dose at<br />

day 28 should be offered to persons who are immunocompromised. Further<br />

doses should be offered to persons who are immunocompromised and have an<br />

inadequate antibody level following the 5th dose of PEP.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

PART 4 VACCINE-PREVENTABLE DISEASES 371<br />

4.16 RABIES AND OTHER<br />

LYSSAVIRUSES (INCLUDING<br />

AUSTRALIAN BAT LYSSAVIRUS)


4.17 ROTAVIRUS<br />

4.17.1 Virology<br />

Rotaviruses are non-enveloped RNA viruses that are classified according to<br />

the two surface proteins they contain: VP7, the ‘G’ glycoprotein, and VP4, the<br />

protease-cleaved ‘P’ protein. <strong>The</strong> G and P proteins are targets for the neutralising<br />

antibodies that contribute to protection against reinfection and disease. 1,2 <strong>The</strong><br />

two gene segments that encode these proteins can segregate independently, and<br />

a binary typing system, consisting of both P and G types, has been developed.<br />

Rotavirus strains are most commonly referred to by their G serotype, with G1,<br />

G2, G3, G4 and G9 accounting for around 90% of serotypes, both globally and in<br />

Australia. 3,4 <strong>The</strong> most common P types found in combination with these G types<br />

are P1A[8] (found with all common G types except G2) and P1B[4], usually found<br />

in combination with G2. 5<br />

4.17.2 Clinical features<br />

Rotavirus is the predominant agent of severe dehydrating gastroenteritis in<br />

infants and young children in both developed and developing countries. 1,2 <strong>The</strong><br />

spectrum of rotavirus infection ranges from asymptomatic infection, to mild,<br />

watery diarrhoea of limited duration, to severe dehydrating diarrhoea with<br />

vomiting, fever, electrolyte imbalance, shock and death. Rotavirus infections are<br />

often more severe than other common causes of diarrhoea, and are more likely to<br />

result in dehydration and hospitalisation. 1,6 <strong>The</strong> incubation period is 1 to 3 days,<br />

after which illness can begin abruptly, with vomiting often preceding the onset<br />

of diarrhoea. 6 Up to one-third of patients have a temperature of >39°C in the first<br />

few days of illness. Symptoms generally resolve in 3 to 7 days.<br />

4.17.3 Epidemiology<br />

Rotaviruses are shed in high concentrations in the stools of infected children<br />

and are transmitted by the faecal–oral route, both through close person-toperson<br />

contact and via fomites. 7 In some instances, rotaviruses might also be<br />

transmitted by other modes, such as faecally contaminated food, water and<br />

respiratory droplets. 6,8<br />

Infection in early childhood is thought to be universal. Although individuals<br />

can be infected several times during their lives, the first infection, typically<br />

between 3 and 36 months of age, is most likely to cause severe diarrhoea and<br />

dehydration. 9,10 <strong>The</strong> degree of protection following natural infection varies. After<br />

a single natural infection, 40% of children are protected against any subsequent<br />

infection with rotavirus, 75% are protected against diarrhoea from a subsequent<br />

rotavirus infection, and 88% are protected against severe diarrhoea. 10 Repeat<br />

infections provide even greater protection. Prior to the introduction of rotavirus<br />

vaccines in Australia, the best available estimates were that approximately<br />

10 000 hospitalisations due to rotavirus occurred each year in children


age group11,12 and affecting 3.8% of all children (1 in 27) by the age of<br />

5 years. In addition to hospitalisations, an estimated 115 000 children 70%) in both rotavirus-specific and all-cause hospital presentations for<br />

gastroenteritis have been reported (Figure 4.17.1). 14-17 Emergency department<br />

visits for acute gastroenteritis have also declined, as have rotavirus<br />

notifications. 18,19<br />

In temperate Australia, rotavirus infections follow a seasonal pattern, with the<br />

peak incidence being in mid to late winter. In the northern tropical and arid<br />

regions, there is no consistent seasonal pattern – disease peaks are unpredictable20 and widespread epidemics cause severe strain on healthcare services. 21,22 Overall,<br />

Indigenous infants and children are hospitalised with rotavirus gastroenteritis<br />

about 3 to 5 times more commonly than their non-Indigenous peers, are younger<br />

at hospitalisation, and have a longer duration of stay (an average of 5 days,<br />

compared with 2 days for non-Indigenous infants). 12,20,21,23<br />

Immunocompromised children and adults, such as those with congenital<br />

immunodeficiency, or post haematopoietic or solid organ transplantation, are at<br />

increased risk of severe, prolonged and even fatal rotavirus gastroenteritis. 1,24,25<br />

Rotavirus is an important cause of nosocomial gastroenteritis, 26-30 and can also<br />

cause disease in adults, especially among those caring for children and those<br />

residing in aged care facilities. 1,31,32<br />

Figure 4.17.1: Rotavirus-coded hospitalisations per month, Australia, 2001 to 2010 17<br />

Number<br />

1200<br />

1000<br />

800<br />

600<br />

400<br />

200<br />

0<br />

Jan-01 Jan-02 Jan-03 Jan-04 Jan-05 Jan-06 Jan-07 Jan-08 Jan-09 Jan-10<br />

Month<br />

National <strong>Immunisation</strong> Program<br />

PART 4 VACCINE-PREVENTABLE DISEASES 373<br />

4.17 ROTAVIRUS


4.17.4 Vaccines<br />

Two oral rotavirus vaccines are available in Australia, and their efficacy and<br />

safety in the prevention of rotavirus gastroenteritis have been extensively<br />

evaluated. 33-39 Both are live attenuated vaccines administered orally to infants,<br />

but the component vaccine viruses differ. <strong>The</strong> human rotavirus vaccine,<br />

Rotarix (GlaxoSmithKline), is a live attenuated vaccine containing one strain<br />

of attenuated human rotavirus (G1P1A[8] strain). Rotarix protects against<br />

non-G1 serotypes on the basis of other shared epitopes. A pentavalent vaccine,<br />

RotaTeq (CSL Limited/Merck & Co Inc), contains five human–bovine rotavirus<br />

reassortants with the human serotypes G1, G2, G3, G4 and P1A[8] and the bovine<br />

serotypes G6 and P7.<br />

In middle- and high-income countries, a course of vaccination with either Rotarix<br />

or RotaTeq prevents rotavirus gastroenteritis of any severity in approximately<br />

70% of recipients and prevents severe rotavirus gastroenteritis and rotavirus<br />

hospitalisation for 85 to 100% of recipients for up to 3 years. 33,34,40,41 Vaccination<br />

is also highly effective in preventing emergency department and clinic/GP<br />

visits. 33,40 Overall, in pre-market clinical trials, rotavirus vaccination prevented<br />

around half (42–58%) of hospital admissions for acute gastroenteritis of any<br />

cause in young children, suggesting that rotavirus is responsible for more<br />

gastroenteritis than detected using routine testing and admission practices. 33,34,40<br />

post-marketing studies in the United States and Australia have confirmed high<br />

vaccine effectiveness and impressive reductions in both rotavirus-coded and allcause<br />

gastroenteritis hospitalisations. 15,19,42-46 Reductions have also been observed<br />

in age groups not eligible for vaccination, suggesting that herd protective effects<br />

are also likely to exist for rotavirus vaccines. 15,19,45<br />

Although more modest estimates of efficacy have been reported in resource-poor<br />

settings, 36-38,47 post-marketing evaluation in the middle income countries Mexico<br />

and Brazil have revealed substantial reductions in diarrhoea-related mortality<br />

since vaccine introduction. 48,49 Studies of Rotarix during consecutive epidemics in<br />

Central Australia gave generally lower and wide-ranging vaccine effectiveness<br />

estimates, which require further investigation. 46,50 Considering the uniqueness<br />

of the remote <strong>Australian</strong> setting, these results should not be extrapolated to<br />

elsewhere in Australia, where the weight of evidence indicates a substantial<br />

reduction in the burden of rotavirus disease following vaccine introduction. To<br />

date, there has been no convincing evidence of important differences between the<br />

two vaccines with regard to protective efficacy against different serotypes. 33,34,39<br />

RotaShield, a tetravalent rhesus-reassortant vaccine, which was licensed in the<br />

United States (but not elsewhere) in 1998–99, was subsequently associated with<br />

intussusception (IS, an uncommon form of bowel obstruction in young children)<br />

in approximately 1 in 10 000 vaccine recipients. 51 <strong>The</strong> pathogenesis of RotaShieldassociated<br />

IS has not been determined. <strong>The</strong> greatest risk of IS occurred within 3<br />

to 14 days after the 1st dose, with a smaller risk after the 2nd dose. 51,52 <strong>The</strong>re is<br />

evidence suggesting that when the 1st dose of RotaShield was given at >3 months<br />

374 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


of age, the risk of IS was increased. 52 <strong>The</strong> current rotavirus vaccines (Rotarix and<br />

RotaTeq) differ in composition to RotaShield, which was more reactogenic. 53-55<br />

<strong>The</strong> large-scale safety studies of Rotarix and RotaTeq included approximately<br />

140 000 infants, and found the risk of IS in vaccine recipients to be similar to that<br />

of placebo recipients, and less than that estimated for RotaShield. 33,34 A metaanalysis<br />

of clinical trial data also did not find evidence of an increased risk of<br />

IS among vaccine recipients. 39 <strong>The</strong> clinical trials of Rotarix and RotaTeq limited<br />

administration of the 1st dose of vaccine to infants under 14 and 12 weeks of age,<br />

respectively, and did not give subsequent doses to infants beyond 24 weeks for<br />

Rotarix and 32 weeks for RotaTeq. 33,34 <strong>The</strong>re are no data from clinical trials on<br />

the use of rotavirus vaccines given outside the recommended dosing age ranges.<br />

While clinical trials excluded an association between Rotarix or RotaTeq and IS<br />

of the magnitude associated with RotaShield, post-marketing studies in Australia<br />

and in Mexico indicate that a smaller increase in the absolute risk of IS might<br />

exist, particularly post dose 1 (see 4.17.11 Adverse events below). 56,57<br />

Vaccine viruses replicate in the intestinal mucosa and can be shed in the stool of<br />

vaccine recipients, particularly after the 1st dose. Vaccine virus shedding is more<br />

common with Rotarix and is detected in the stool a week after vaccination in up<br />

to 80% of 1st dose recipients, and in up to 30% of 2nd dose recipients. 58,59 RotaTeq<br />

is only shed after the 1st dose (in up to 13% of recipients). 33 In one study of 80<br />

sets of twins, transmission of Rotarix was observed to occur from 15 vaccinated<br />

infants to their unvaccinated twin, 60 indicating that transmission of vaccine virus<br />

to unvaccinated contacts is likely to occur, but the clinical implication of this has<br />

not been studied (see 4.17.10 Precautions below).<br />

Adventitious DNA fragments of porcine circoviruses have been detected in both<br />

Rotarix and RotaTeq vaccines. However, porcine circoviruses have never been<br />

shown to cause illness in humans and are considered non-pathogenic.<br />

• Rotarix – GlaxoSmithKline (live attenuated RIX4414 human rotavirus<br />

strain, type G1P1A[8]). Each 1.5 mL monodose pre-filled oral<br />

applicator or squeezable tube contains ≥10 6.0 cell culture infectious<br />

dose 50% (CCID 50 ) of the RIX4414 strain; di-sodium adipate;<br />

Dulbecco’s Modified Eagle Medium; sterile water. Manufacture<br />

involves exposure to bovine-derived material.<br />

• RotaTeq – CSL Limited/Merck & Co Inc (live attenuated human–<br />

bovine reassortant rotavirus strains, types G1, G2, G3, G4 and<br />

P1A[8]). Each 2.0 mL monodose pre-filled dosing tube contains a<br />

minimum dose level of at least 2.0 x 10 6 infectious units of each of<br />

the rotavirus reassortants G1, G2, G3, G4 and P1A[8]; sodium citrate;<br />

sodium phosphate monobasic monohydrate; sodium hydroxide;<br />

polysorbate 80; cell culture medium. Manufacture involves exposure<br />

to bovine-derived material.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 375<br />

4.17 ROTAVIRUS


4.17.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 61 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

4.17.6 Dosage and administration<br />

Rotavirus vaccines are for oral administration only. Under no circumstances<br />

should rotavirus vaccines be injected.<br />

Rotarix is recommended for use in a 2-dose course in infants and upper age<br />

limits apply; see Table 4.17.1. <strong>The</strong> liquid formulation is presented as a clear,<br />

colourless liquid contained within an oral applicator (syringe-type applicator<br />

with a plunger stopper or a squeezable tube). <strong>The</strong> 1.5 mL dose of vaccine should<br />

be administered orally from the oral applicator onto the inside of the infant’s<br />

cheek. Rotarix does not require reconstitution or dilution.<br />

RotaTeq is recommended for use in a 3-dose course in infants and upper<br />

age limits apply; see Table 4.17.1. It is supplied in a container consisting of a<br />

squeezable plastic, latex-free dosing tube with a twist-off cap, allowing for direct<br />

oral administration of the 2 mL dose onto the inside of the infant’s cheek. RotaTeq<br />

does not require reconstitution or dilution. RotaTeq is a pale yellow, clear liquid<br />

that may have a pink tint.<br />

<strong>The</strong>re are limited data available on the safety of administering higher than<br />

the recommended dose of rotavirus vaccines or the efficacy of a partially<br />

administered dose(s). If most of an oral rotavirus vaccine dose has been spat<br />

out or vomited within minutes of administration, a single repeat dose can be<br />

administered during the same visit. If an infant regurgitates or vomits only a<br />

small part of a vaccine dose, it is not necessary to repeat the dose. <strong>The</strong>refore, the<br />

regurgitated (and incomplete volume) dose is still considered as the valid dose.<br />

Co-administration with other vaccines<br />

Rotavirus vaccines can be co-administered with other vaccines included on the<br />

NIP schedule at 2 and 4 months of age (Rotarix) or 2, 4 and 6 months of age<br />

(RotaTeq). <strong>The</strong> available evidence from clinical trials suggests co-administration<br />

of oral rotavirus vaccines is safe and effective and does not interfere with<br />

the immune response to other vaccine antigens (DTPa, Hib, IPV, hepB, and<br />

pneumococcal conjugate vaccines). 58,59,62<br />

<strong>The</strong>re are no restrictions on the timing of administration of any other live<br />

vaccines in relation to rotavirus vaccines, including BCG or oral poliomyelitis<br />

vaccine (OPV), for example, in infants who have received OPV overseas. Delay of<br />

rotavirus vaccination for 4 weeks following vaccination with BCG or vice versa is<br />

not necessary.<br />

376 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Interchangeability of rotavirus vaccines<br />

Completion of a course of rotavirus vaccine should be with vaccine from the<br />

same manufacturer whenever possible. <strong>The</strong>re are no studies that address the<br />

interchangeability of the two available rotavirus vaccines. However, if either<br />

dose 1 or 2 of vaccine is given as RotaTeq, a 3rd dose of either rotavirus vaccine<br />

should be given, provided that the upper age limit and inter-vaccine interval, as<br />

defined in Table 4.17.1, are met.<br />

4.17.7 Recommendations<br />

Infants<br />

Administration of a course of oral rotavirus vaccination is recommended for<br />

all infants in the first half of the 1st year of life. Vaccination of older infants and<br />

children is not recommended as there are theoretical concerns regarding use in<br />

older age groups (see 4.17.4 Vaccines above). Vaccination should occur at either 2<br />

and 4 months of age (Rotarix), or 2, 4 and 6 months of age (RotaTeq), according<br />

to the following schedules and upper age limits (see Table 4.17.1). <strong>The</strong> 1st dose of<br />

either rotavirus vaccine can be given as early as 6 weeks of age, where necessary<br />

(see Table 4.17.1). If the 1st dose is given at 6 weeks of age, the next scheduled<br />

rotavirus vaccine dose(s) should still be given according to the age limits<br />

specified for dosing in Table 4.17.1 below.<br />

Rotarix (human monovalent rotavirus vaccine)<br />

<strong>The</strong> vaccination course of Rotarix consists of 2 doses, at 2 and 4 months of age.<br />

<strong>The</strong> 1st dose should be given between 6 and 14 weeks of age (i.e. prior to turning<br />

15 weeks old), and the 2nd dose should be given by 24 weeks of age (i.e. prior to<br />

turning 25 weeks old). <strong>The</strong> interval between the 2 doses should not be less than<br />

4 weeks.<br />

RotaTeq (pentavalent human–bovine reassortant rotavirus vaccine)<br />

<strong>The</strong> vaccination course of RotaTeq consists of 3 doses, at 2, 4, and 6 months of age.<br />

<strong>The</strong> 1st dose should be given between 6 and 12 weeks of age (i.e. prior to turning<br />

13 weeks old), and all doses should be given by 32 weeks of age (i.e. prior to<br />

turning 33 weeks old). <strong>The</strong> interval between doses should be at least 4 weeks.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 377<br />

4.17 ROTAVIRUS


Table 4.17.1: Upper age limits for dosing of oral rotavirus vaccines<br />

Rotarix<br />

(GlaxoSmithKline)<br />

RotaTeq (CSL<br />

Limited/Merck<br />

& Co Inc)<br />

Doses Age of<br />

routine oral<br />

administration<br />

2 oral doses<br />

(1.5 mL/dose)<br />

3 oral doses<br />

(2 mL/dose)<br />

2 and<br />

4 months<br />

2, 4 and<br />

6 months<br />

378 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Recommended age limits<br />

for dosing<br />

1st<br />

dose<br />

6–14*<br />

weeks<br />

6–12 †<br />

weeks<br />

2nd<br />

dose<br />

10–24*<br />

weeks<br />

10–32 †<br />

weeks<br />

3rd<br />

dose<br />

Minimum<br />

interval<br />

between<br />

doses<br />

N/A 4 weeks<br />

14–32 †<br />

weeks<br />

4 weeks<br />

* <strong>The</strong> upper age limit for receipt of the 1st dose of Rotarix is immediately prior to turning<br />

15 weeks old, and the upper age limit for receipt of the 2nd dose is immediately prior to turning<br />

25 weeks old.<br />

† <strong>The</strong> upper age limit for receipt of the 1st dose of RotaTeq is immediately prior to turning<br />

13 weeks old. <strong>The</strong> 2nd dose of vaccine should preferably be given by 28 weeks of age to allow<br />

for a minimum interval of 4 weeks before receipt of the 3rd dose. <strong>The</strong> upper age limit for the 3rd<br />

dose is immediately prior to turning 33 weeks old. For infants presenting for their 2nd dose after<br />

reaching 29 weeks of age, a 2nd and final dose can be given, provided the upper age limit of<br />

32 weeks (immediately prior to turning 33 weeks old) has not been reached.<br />

For infants in whom the 1st dose of rotavirus vaccine is inadvertently<br />

administered at an age greater than the suggested cut-off (i.e. after the 14th week<br />

of age for Rotarix or the 12th week of age for RotaTeq), the remaining vaccine<br />

doses should be administered as per the schedule, providing the minimum<br />

interval between doses can be maintained within the recommended age limits<br />

for subsequent doses. <strong>The</strong> timing of the 1st dose should not affect the safety and<br />

efficacy of the 2nd and 3rd doses. 6 Infants who develop rotavirus gastroenteritis<br />

before receiving the full course of rotavirus vaccination should still complete<br />

the full 2- or 3-dose schedule (dependent on the brand of vaccine), because one<br />

rotavirus infection only provides partial immunity. 6<br />

Older infants<br />

Vaccination of older infants, children or adults is not recommended. Infants<br />

should commence the course of rotavirus vaccination within the recommended<br />

age limits for the 1st dose and doses should not be given beyond the upper age<br />

limits for the final dose of the vaccine course (see ‘Infants’ above). <strong>The</strong> incidence<br />

of severe rotavirus infection decreases with increasing age and the benefit and<br />

safety profile of rotavirus vaccination in older infants and children has not been<br />

established.<br />

Preterm infants<br />

Vaccination of preterm infants using either rotavirus vaccine is indicated at a<br />

chronologic age (without correction for prematurity) of at least 6 weeks, if the<br />

infant is clinically stable. Preterm infants (born at


gestational age; median 34 weeks) who experienced rates of adverse events after<br />

vaccination similar to matched placebo recipients. 6 Efficacy against rotavirus<br />

gastroenteritis of any severity appeared comparable to efficacy in full-term<br />

infants (73%; 95% CI: –2 to 95%). 64 <strong>The</strong>se conclusions would also be expected<br />

to apply to Rotarix vaccine, which appears safe and immunogenic in preterm<br />

infants. 65 If standard infection control precautions are maintained, administration<br />

of rotavirus vaccine to hospitalised infants, including hospitalised preterm<br />

infants, would be expected to carry a low risk for transmission of vaccine viruses.<br />

See also 4.17.10 Precautions below for other special risk groups and hospitalised<br />

infants.<br />

4.17.8 Pregnancy and breastfeeding<br />

<strong>The</strong>re are no restrictions on the infant’s consumption of food or liquid, including<br />

breast milk, either before or after vaccination with either rotavirus vaccine. 6,62<br />

Infants living in households of pregnant women can receive rotavirus vaccines.<br />

Most pregnant women will have pre-existing immunity to rotavirus, but<br />

protection from transmission of wild-type infection through the vaccination of<br />

infant contacts may benefit adults, including pregnant women, and outweighs<br />

any theoretical concern regarding exposure to vaccine viruses.<br />

4.17.9 Contraindications<br />

<strong>The</strong> contraindications to rotavirus vaccines are:<br />

• anaphylaxis following a previous dose of either rotavirus vaccine<br />

• anaphylaxis following any vaccine component<br />

• previous history of intussusception or a congenital abnormality that may<br />

predispose to IS<br />

<strong>The</strong> risk of recurrence of IS unrelated to rotavirus vaccination is in the order<br />

of 10%. 66 In addition, certain congenital malformations affecting the gut<br />

(e.g. Meckel’s diverticulum) increase the risk of IS. Because of the possible<br />

association of rotavirus vaccination with an increased risk of IS (see 4.17.11<br />

Adverse events below), it is considered prudent to withhold administration of<br />

rotavirus vaccines to an infant with a previous history of IS or with a known<br />

uncorrected congenital malformation associated with increased risk of IS.<br />

• severe combined immunodeficiency (SCID) in infants<br />

Case reports from the United States67-69 indicate prolonged vaccine virusassociated<br />

gastrointestinal disease following receipt of rotavirus vaccines<br />

among infants with SCID. Because these infants are unlikely to generate a<br />

protective immune response to vaccination and because of potential harm,<br />

rotavirus vaccines are contraindicated for infants with SCID. For infants<br />

with less severe forms of immunocompromise, the risk of vaccine-associated<br />

disease is likely to be less than the risk of natural infection (see 4.17.10<br />

Precautions below).<br />

PART 4 VACCINE-PREVENTABLE DISEASES 379<br />

4.17 ROTAVIRUS


4.17.10 Precautions<br />

Infants with acute gastroenteritis<br />

Infants with moderate to severe acute gastroenteritis should not be vaccinated<br />

until after recovery from their acute illness. Infants with mild gastroenteritis<br />

(including mild diarrhoea) can be vaccinated. <strong>The</strong> use of rotavirus vaccines has<br />

not been studied in infants with acute gastroenteritis.<br />

Infants with moderate to severe illness<br />

As with other vaccines, infants with a moderate to severe illness should be<br />

vaccinated after recovery. In addition to the factors mentioned above, this<br />

avoids superimposing potential adverse events related to vaccination with the<br />

concurrent illness.<br />

Infants with underlying conditions predisposing to severe rotavirus<br />

gastroenteritis<br />

Conditions predisposing to severe or complicated rotavirus gastroenteritis<br />

include metabolic disorders and chronic gastrointestinal disease, such as<br />

Hirschsprung’s disease, malabsorption syndromes or short gut syndrome. 1<br />

Data on the safety of live rotavirus vaccines among such infants is limited. In<br />

one report, RotaTeq was reported to be tolerated in 8 of 9 infants with highoutput<br />

ileostomies, while 1 infant experienced an increase in ileostomy losses. 70<br />

However, because of the greater risk of serious rotavirus disease, the benefits<br />

from vaccination are expected to outweigh the risk in these infants.<br />

Infants who are immunocompromised<br />

<strong>The</strong>re are theoretical concerns that vaccine-associated gastrointestinal disease<br />

could occur in immunocompromised infants who receive rotavirus vaccines, and<br />

infants with the most severe forms of immunocompromise (SCID) should not<br />

receive rotavirus vaccine (see 4.17.9 Contraindications above). However, the risk<br />

for those infants with less severe immunocompromise may be less than the risk<br />

from natural infection. <strong>The</strong> risks and benefits of vaccination should be considered<br />

in the context of the infant’s specific immunocompromise with appropriate<br />

specialist advice6 (see 3.3.3 Vaccination of immunocompromised persons).<br />

Rotavirus vaccines have been administered to HIV-infected infants in clinical<br />

trial settings. 36-38 Specific data on the safety and efficacy of rotavirus vaccines in<br />

these infants are limited, but suggest that the vaccines are safe and immunogenic<br />

in HIV-infected, but clinically stable, children. 71,72 (See also 3.3.3 Vaccination<br />

of immunocompromised persons and Table 3.3.4 Categories of immunocompromise<br />

in HIV-infected persons, based on age-specific CD4 + counts and percentage of total<br />

lymphocytes.) <strong>The</strong>re are no data on the use of rotavirus vaccines in infants born to<br />

women who have received immunosuppressive therapy in pregnancy (see ‘Use<br />

of immunosuppressive therapy during pregnancy’ in 3.3.2 Vaccination of women<br />

who are planning pregnancy, pregnant or breastfeeding, and preterm infants).<br />

380 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Infants living in households with people who are immunocompromised<br />

Infants living in households with immunocompromised persons should be<br />

vaccinated. In general, immunocompromised household members are afforded<br />

protection by vaccination of young children in the household and this is<br />

considered to outweigh the risk of transmitting vaccine virus shed in stools to the<br />

immunocompromised household member. However, there have been no studies<br />

to specifically address this question. 6 Hand washing and the careful disposal of<br />

soiled nappies are likely to minimise any risk of vaccine transmission to other<br />

household members. (See also 3.3.3 Vaccination of immunocompromised persons.)<br />

Recent administration of antibody-containing blood products<br />

Infants who have recently received antibody-containing blood products and are<br />

at an eligible age should be vaccinated. <strong>The</strong> interval between vaccination and<br />

receipt of the blood product should be as long as possible, but without delaying<br />

administration of vaccine beyond the suggested age limits for dosing (as per<br />

Table 4.17.1 above). This recommendation for maximising the interval between<br />

receipt of antibody-containing blood products and rotavirus vaccination is based<br />

on theoretical concern that passively acquired antibody to rotavirus may interfere<br />

with vaccine immunogenicity. 6<br />

Hospitalised infants<br />

Administration of rotavirus vaccine to hospitalised infants, including premature<br />

infants, is likely to carry a low risk for transmission of vaccine viruses if<br />

standard infection control precautions are maintained. Provided that the infant<br />

is medically stable, vaccination should not be delayed, particularly if the delay<br />

would result in an infant being beyond the upper age limit for vaccination (see<br />

4.17.7 Recommendations above). If a recently vaccinated child is hospitalised for<br />

any reason, no precautions other than routine standard precautions need be<br />

taken to prevent the spread of vaccine virus in the hospital setting.<br />

4.17.11 Adverse events<br />

Intussusception<br />

Although clinical trials of the two available vaccines did not find an association<br />

between vaccination and intussusception (IS) 39 (see 4.17.4 Vaccines above), one<br />

post-marketing study in Australia found evidence of a 4- to 5-fold increase in<br />

the risk of IS in the 7 days following the 1st dose of either Rotarix or RotaTeq. 56<br />

However, no overall increase in the risk of IS was detectable over the first<br />

9 months of life. 56 A similar apparent increase in risk for IS following the 1st dose<br />

of Rotarix has been observed in Mexico, and a smaller increase after the 2nd dose<br />

of Rotarix in Brazil. 57 A study in the United States found no increased risk of IS<br />

following RotaTeq; however, the study was limited by small numbers, which<br />

reduced power to determine a low range risk increase. 73 A subsequent <strong>Australian</strong><br />

study estimated the increased risk of IS to be approximately 9-fold in the first<br />

PART 4 VACCINE-PREVENTABLE DISEASES 381<br />

4.17 ROTAVIRUS


7 days after dose 1, and 2-fold in the first 7 days after dose 2 of either vaccine. 74<br />

<strong>The</strong> baseline risk of intussusception for <strong>Australian</strong> infants is around 80 cases per<br />

100 000 infants. 75 <strong>The</strong> increased risk of IS following rotavirus vaccination, from<br />

the most recent <strong>Australian</strong> study, is estimated as approximately 6 additional<br />

cases of intussusception among every 100 000 infants vaccinated, or 18 additional<br />

cases per year in Australia. 74 This estimate assumes that infants in which<br />

an episode of IS occurs shortly after vaccination would not have otherwise<br />

experienced a ‘natural’ episode of intussusception; however, this cannot be<br />

determined from current data. Importantly, studies from both Australia14-17 and<br />

overseas57 have demonstrated the substantial impact of vaccination in preventing<br />

rotavirus morbidity and mortality (see also 4.17.3 Epidemiology above). Rotavirus<br />

vaccines continue to be recommended for use on the basis of this positive benefit<br />

to risk profile. 76 <strong>Immunisation</strong> providers should inform parents and carers of the<br />

rare risk of intussusception and how to be alert for the signs and symptoms of<br />

the condition.<br />

Rotavirus vaccine should not be given to an infant who has had a confirmed<br />

intussusception because there may be an increased risk of the condition recurring<br />

(see 4.17.9 Contraindications above).<br />

Other adverse events<br />

No significant increase in post-vaccination vomiting, diarrhoea or fever has<br />

been reported during follow-up of several thousand recipients of Rotarix<br />

compared to those who were unvaccinated. 39,77 Detailed follow-up of 11 700<br />

recipients of RotaTeq or placebo reported no increase in fever or irritability in<br />

the week after vaccination among vaccinated infants, but a small increase in<br />

the incidence of vomiting (7% versus 5%) and diarrhoea (10% versus 9%). 78<br />

Vomiting and diarrhoea have not emerged as important adverse events following<br />

immunisation in post-marketing surveillance of rotavirus vaccines.<br />

Infants who report an episode of diarrhoea or vomiting following vaccination<br />

should still receive subsequent rotavirus vaccine doses, as required and age<br />

eligible. <strong>The</strong> potential causes of diarrhoea/vomiting following vaccination<br />

include: gastroenteritis unrelated to rotavirus vaccination or infection (e.g.<br />

another viral agent); natural rotavirus infection (as vaccination is neither<br />

immediately protective nor 100% protective against all disease); or symptoms<br />

from vaccine virus replication (less likely). If rotavirus is detected by routine stool<br />

testing on a recently vaccinated infant, a positive test result can represent either<br />

natural infection or vaccine virus (as vaccine virus shedding occurs commonly<br />

after vaccination (see 4.17.4 Vaccines above). Specific testing is required to<br />

differentiate between vaccine virus and natural infection; however, this is rarely<br />

clinically indicated.<br />

382 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.17.12 Variations from product information<br />

<strong>The</strong> product information for Rotarix states that the vaccine should not be<br />

administered to subjects with any chronic gastrointestinal disease. <strong>The</strong> ATAGI<br />

recommends instead that pre-existing chronic gastrointestinal disease is not a<br />

contraindication to rotavirus vaccination, with the exception of those conditions<br />

that may predispose to IS (see 4.17.9 Contraindications and 4.17.10 Precautions<br />

above).<br />

<strong>The</strong> product information for RotaTeq states that in the event that a dose of<br />

vaccine is spat out or vomited post vaccination, a replacement dose should not<br />

be given. <strong>The</strong> ATAGI recommends instead that if most of a dose is spat out or<br />

vomited then a single replacement dose may be given (see 4.17.6 Dosage and<br />

administration above.)<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

PART 4 VACCINE-PREVENTABLE DISEASES 383<br />

4.17 ROTAVIRUS


4.18 RUBELLA<br />

4.18.1 Virology<br />

Rubella is an enveloped togavirus, genus Rubivirus. <strong>The</strong> virus has an RNA<br />

genome and is closely related to group A arboviruses, but does not require<br />

a vector for transmission. It is relatively unstable, and is inactivated by lipid<br />

solvents, trypsin, formalin, extremes of heat and pH, and light. 1<br />

4.18.2 Clinical features<br />

Rubella is generally a mild and self-limiting infectious disease, spread<br />

from person to person by respiratory secretions, possibly including aerosol<br />

transmission. 1,2 It causes a transient, generalised, erythematous, maculopapular<br />

rash; lymphadenopathy involving the post-auricular and sub-occipital glands;<br />

and, occasionally, arthritis and arthralgia. Other complications, such as<br />

neurological disorders and thrombocytopenia, may occur but are rare. Clinical<br />

diagnosis is unreliable since the symptoms are often fleeting and can be caused<br />

by other viruses; in particular, the rash is not unique to rubella and may be<br />

absent. 1,2 Up to 50% of rubella virus infections are subclinical or asymptomatic. 1<br />

A history of rubella should, therefore, not be accepted without serological<br />

evidence of previous infection. 1 <strong>The</strong> incubation period is 14 to 21 days, and the<br />

period of infectivity is from 1 week before until 4 days after the onset of the rash. 2<br />

Rubella infection in pregnancy can result in fetal infection, causing congenital<br />

rubella syndrome (CRS) in a high proportion of cases. Up to 90% of infants<br />

born to women who had rubella infection in the first trimester of pregnancy<br />

have abnormalities (often multiple) characteristic of CRS. 3-5 <strong>The</strong> risk of damage<br />

declines to 10 to 20% by 16 weeks gestation. After this stage of pregnancy,<br />

fetal damage is rare but has been reported up to 20 weeks gestation. 3 <strong>The</strong><br />

characteristics of CRS include intellectual disabilities, cataracts, deafness, cardiac<br />

abnormalities, intrauterine growth retardation, and inflammatory lesions of<br />

the brain, liver, lungs and bone marrow. 3 Any combination of these defects<br />

may occur, but defects that commonly occur alone following infection after<br />

the first 8 weeks of pregnancy are deafness and pigmentary retinopathy. Some<br />

infected infants may appear normal at birth, but defects, especially sensorineural<br />

deafness, may be detected later. 6<br />

Rubella infection has been reported in some persons who already have either<br />

natural or vaccine-induced antibody. 3 Occasional cases of CRS after reinfection in<br />

pregnancy have been documented. However, fetal damage is very rare in cases of<br />

infection in women in whom antibody has previously been detected. 4,7-9<br />

4.18.3 Epidemiology<br />

Evidence suggests that endemic rubella is well controlled in Australia. 10 <strong>The</strong><br />

incidence of rubella has fallen rapidly since vaccine registration, and notifications<br />

of rubella have been low since high vaccine coverage was achieved with the<br />

National Measles Control Campaign in late 1998 and then maintained. 10 Since<br />

384 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


2003, rubella notifications in Australia have been less than 0.3 per 100 000. <strong>The</strong>re<br />

has been a shift in the age distribution of cases, with comparatively more cases<br />

seen in older age groups, particularly the 25–29 years age group. 10<br />

Rubella vaccines have been registered in Australia since 1970, and mass<br />

vaccination of schoolgirls commenced in 1971. 1,11 Non-pregnant, seronegative<br />

adult women were also vaccinated. <strong>The</strong>se programs were successful and there<br />

was a significant reduction in the incidence of CRS from 1977. 12-14 Successful<br />

vaccination campaigns and high vaccination coverage resulted in no cases of<br />

congenital rubella syndrome occurring in infants of <strong>Australian</strong>-born mothers<br />

between 1998 and 2002. However, 5 cases resulting from infection acquired<br />

outside of Australia were reported during this time. 15 In 2003, 2 cases of CRS<br />

occurred in <strong>Australian</strong>-born mothers from infection that occurred in Australia, 16<br />

which reinforces the need for high vaccination coverage of women of childbearing<br />

age. Between 2004 and 2008, 2 confirmed cases of CRS were reported in<br />

Australia, in children whose mothers were born outside Australia. 17-19<br />

<strong>The</strong>re has also been a significant increase in the percentage of pregnant women<br />

immune to rubella (e.g. in New South Wales from 82% in 1971 to 96% in 1983). 20<br />

Based on a study conducted in Melbourne in 2000, it was estimated that only<br />

2.5% of women of child-bearing age in Australia were seronegative. 21 However,<br />

susceptibility was higher among certain groups of women, particularly overseasborn<br />

women (see ‘Women of child-bearing age, including post-partum women’<br />

in 4.18.7 Recommendations below). 21<br />

Young adult males may not be immune to rubella, because they did not receive<br />

a measles-mumps-rubella (MMR) vaccine. 22 <strong>The</strong> MMR vaccination program for<br />

all adolescents replaced the rubella program for girls in 1993/94. 11 A serosurvey<br />

conducted in 1999 showed that only 84% of males aged 14–18 years (compared to<br />

95% of females) and 89% of males aged 19–49 years (compared to 98% of females)<br />

were immune to rubella. 22 For this reason, young adult males, as well as females,<br />

who do not have a documented history of receipt of 2 doses of MMR vaccine<br />

should be vaccinated (see 4.18.7 Recommendations below). This is both for their<br />

own protection and to prevent transmission of the infection in the community.<br />

Goals for the elimination of rubella and CRS have been set by a number of<br />

World Health Organization (WHO) regions, and elimination has been declared<br />

by the Pan American Health Organization. 23 <strong>The</strong> WHO Western Pacific Region<br />

has set goals for increased rubella and CRS control efforts, with a number<br />

of member states yet to incorporate rubella vaccination into their routine<br />

schedule. 24 As with elimination of measles, rubella and CRS elimination<br />

requires continued strengthening of immunisation and surveillance efforts,<br />

particularly identification of rubella virus genotypes to confirm the absence of<br />

an endemic strain. 25<br />

PART 4 VACCINE-PREVENTABLE DISEASES 385<br />

4.18 RUBELLA


4.18.4 Vaccines<br />

Monovalent rubella vaccine is not available in Australia. Rubella vaccination<br />

is provided using either MMR or measles-mumps-rubella-varicella (MMRV)<br />

vaccines. Two quadrivalent combination vaccines containing live attenuated<br />

measles, mumps, rubella and varicella viruses (MMRV) are registered in Australia.<br />

A single dose of rubella vaccine produces an antibody response in over 95% of<br />

vaccine recipients, but antibody levels are lower than after natural infection. 3,7,8<br />

A 2nd dose aims to confer immunity in those who fail to seroconvert to the 1st<br />

dose. Vaccine-induced antibodies have been shown to persist for at least 16 years<br />

in the absence of endemic disease. 7,8,26,27 Protection against clinical rubella appears<br />

to be long-term in those who seroconvert. 3<br />

Combination MMRV vaccines have been shown, in clinical trials, to produce<br />

similar rates of seroconversion to all four vaccine components compared with<br />

MMR and monovalent varicella vaccines administered concomitantly at separate<br />

injection sites. 28-31<br />

Trivalent measles–mumps–rubella (MMR) vaccines<br />

• M-M-R II – CSL Limited/Merck & Co Inc (live attenuated measles<br />

virus [Enders’ attenuated Edmonston strain], mumps virus [Jeryl<br />

Lynn B level strain] and rubella virus [Wistar RA 27/3 strain]).<br />

Lyophilised pellet in a monodose vial with separate diluent. Each<br />

0.5 mL reconstituted dose contains ≥1000 tissue culture infectious<br />

dose 50% (TCID ) of Enders’ attenuated Edmonston measles<br />

50<br />

virus, ≥12 500 TCID of the Jeryl Lynn B level mumps virus, and<br />

50<br />

≥1000 TCID of the Wistar RA 27/3 rubella virus; sorbitol; sucrose;<br />

50<br />

hydrolysed gelatin; human albumin; fetal bovine serum; neomycin.<br />

• Priorix – GlaxoSmithKline (live attenuated measles virus [Schwarz<br />

strain], mumps virus [RIT 4385 strain, derived from the Jeryl Lynn<br />

strain] and rubella virus [Wistar RA 27/3 strain]). Lyophilised pellet<br />

in a monodose vial with a pre-filled diluent syringe. Each 0.5 mL<br />

reconstituted dose contains ≥10 3.0 cell culture infectious dose 50%<br />

(CCID 50 ) of the Schwarz measles virus, ≥10 3.7 CCID 50 of the RIT 4385<br />

mumps virus, and ≥10 3.0 CCID 50 of the Wistar RA 27/3 rubella virus;<br />

lactose; neomycin; sorbitol; mannitol.<br />

Quadrivalent measles-mumps-rubella-varicella (MMRV) vaccines<br />

• Priorix-tetra – GlaxoSmithKline (live attenuated measles virus<br />

[Schwarz strain], mumps virus [RIT 4385 strain, derived from the<br />

Jeryl Lynn strain], rubella virus [Wistar RA 27/3 strain] and varicella-<br />

386 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


zoster virus [Oka strain]). Lyophilised pellet in a monodose vial with<br />

a pre-filled diluent syringe. Each 0.5 mL reconstituted dose contains<br />

≥10 3.0 CCID 50 of the Schwarz measles virus, ≥10 4.4 CCID 50 of the RIT<br />

4385 mumps virus, ≥10 3.0 CCID 50 of the Wistar RA 27/3 rubella virus,<br />

and ≥10 3.3 plaque-forming units (PFU) of Oka varicella-zoster virus;<br />

lactose; neomycin; sorbitol; mannitol.<br />

• ProQuad – CSL Limited/Merck & Co Inc (live attenuated measles<br />

virus [Enders’ attenuated Edmonston strain], mumps virus [Jeryl<br />

Lynn B level strain], rubella virus [Wistar RA 27/3 strain] and<br />

varicella-zoster virus [Oka/Merck strain]). Lyophilised powder<br />

in a monodose vial with a pre-filled diluent syringe. Each 0.5 mL<br />

reconstituted dose contains ≥103.0 TCID of Enders’ attenuated<br />

50<br />

Edmonston measles virus, ≥104.3 TCID of the Jeryl Lynn B level<br />

50<br />

mumps virus, ≥103.0 TCID of the Wistar RA 27/3 rubella virus, and<br />

50<br />

≥103.99 PFU of Oka/Merck varicella virus; sucrose; hydrolysed gelatin;<br />

urea; sorbitol; monosodium L-glutamate; human albumin; neomycin;<br />

residual components of MRC-5 cells; bovine serum albumin.<br />

4.18.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 32 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

Both MMR and MMRV vaccines must be reconstituted by adding the entire<br />

contents of the diluent container to the vial containing the pellet and shaking<br />

until the pellet is completely dissolved.<br />

Reconstituted Priorix (MMR), M-M-R II (MMR) and Priorix-tetra (MMRV)<br />

vaccines should be used as soon as practicable. If storage is necessary, hold at<br />

+2°C to +8°C for not more than 8 hours.<br />

Reconstituted ProQuad (MMRV) vaccine must be used within 30 minutes.<br />

4.18.6 Dosage and administration<br />

<strong>The</strong> dose of Priorix (MMR) vaccine for both children and adults is 0.5 mL, to be<br />

given by either SC or IM injection.<br />

<strong>The</strong> dose of M-M-R II (MMR) vaccine for both children and adults is 0.5 mL, to<br />

be given by SC injection.<br />

For children


Co-administration with other vaccines<br />

MMR or MMRV vaccines can be given at the same time as other live attenuated<br />

parenteral vaccines (e.g. varicella, BCG, yellow fever) or other inactivated<br />

vaccines (including DTPa, hepatitis B, Hib, IPV, MenCCV, hepatitis A and<br />

pneumococcal conjugate vaccine), 34 using separate syringes and injection sites.<br />

If MMR or MMRV vaccine is not given simultaneously with other live attenuated<br />

parenteral vaccines, they should be given at least 4 weeks apart.<br />

If MMR vaccine is given at the same time as monovalent varicella vaccine (VV),<br />

they should be given using separate syringes and injection sites. MMR vaccine<br />

and monovalent VV should not be mixed together prior to injection.<br />

Separate administration of measles, mumps or rubella vaccine is not available<br />

as an alternative to MMR vaccine, although a monovalent varicella vaccine is<br />

available (see 4.22 Varicella).<br />

Interchangeability of MMR-containing vaccines<br />

In general, the two brands of MMR vaccine can be considered interchangeable,<br />

that is, the 2nd MMR dose does not have to be of the same brand as the 1st. <strong>The</strong><br />

same principle applies to the two available MMRV vaccines, 34 although they are<br />

not routinely recommended in a 2-dose schedule.<br />

4.18.7 Recommendations<br />

For further information on the recommendations for MMR and MMRV vaccines,<br />

see 4.9 Measles and 4.22 Varicella.<br />

<strong>The</strong> principal aim of rubella vaccination is to prevent congenital rubella<br />

syndrome by stopping the circulation of rubella virus in the community.<br />

Susceptible pregnant women will continue to be at risk of rubella infection in<br />

pregnancy until the transmission of rubella virus is interrupted by continued<br />

high-level coverage of rubella-containing vaccine.<br />

A history of rubella is not a contraindication to vaccination. Persons who<br />

are already immune to rubella have no increased risk of side effects from<br />

vaccination. 3,7<br />

Infants aged


children


avoid the risk of transmitting rubella to pregnant women 35 (see 3.3 Groups with<br />

special vaccination requirements, Table 3.3.7 Recommended vaccinations for persons at<br />

increased risk of certain occupationally acquired vaccine-preventable diseases).<br />

Women of child-bearing age, including post-partum women<br />

Every effort should be made to identify and immunise non-pregnant<br />

seronegative women of child-bearing age (see ‘Serological testing for immunity<br />

to rubella’ below). <strong>The</strong> following women are more likely to be seronegative to<br />

rubella: women born overseas (especially in Asia, Pacific islands, sub-Saharan<br />

Africa and South America) who entered Australia after the age of routine<br />

vaccination; Indigenous women living in rural and remote regions; non-English<br />

speaking women; women over the age of 35 years; and <strong>Australian</strong>-born Muslim<br />

women. 12,13,21,36-38<br />

Seronegative women should be given MMR vaccine and advised to avoid<br />

pregnancy for 28 days after vaccination. Vaccinated women should be tested<br />

for seroconversion 6 to 8 weeks after vaccination (see ‘Serological testing for<br />

immunity to rubella’ below). Women who have negative or very low antibody<br />

levels after vaccination should be revaccinated. However, if antibody levels<br />

remain low after a 2nd documented vaccination, it is unlikely that further<br />

vaccinations will improve this. 3 Further testing and vaccination is not usually<br />

warranted; however, consultation with the laboratory that performed the<br />

serological testing may also be helpful (see also ‘Serological testing for immunity<br />

to rubella’ below). Negative serology after 2 documented doses of rubellacontaining<br />

vaccine may represent a false negative (i.e. an antibody titre too low<br />

to be detected using routine commercial assays).<br />

Although 2 doses of MMR vaccine are routinely recommended, if rubella<br />

immunity is demonstrated after receipt of 1 dose of a rubella-containing vaccine,<br />

no further dose is required, unless indicated by subsequent serological testing<br />

(see ‘Serological testing for immunity to rubella’ below) or if indicated for<br />

protection against measles and mumps (see 4.9 Measles and 4.11 Mumps).<br />

Women found to be seronegative on antenatal testing for rubella immunity<br />

should be vaccinated after delivery and before discharge from the maternity<br />

unit, as discussed above. <strong>The</strong>se women should be tested for rubella immunity 6<br />

to 8 weeks following vaccination. 1,7 (See also ‘Serological testing for immunity to<br />

rubella’ below.)<br />

Serological testing for immunity to rubella<br />

Serological testing for immunity to rubella after routine vaccination of children<br />

is not recommended. However, serological testing for rubella immunity can be<br />

performed in cases where a history of natural immunity or 2 doses of vaccine<br />

administration is uncertain. It is particularly important to ensure that women<br />

of child-bearing age are immune to rubella (see ‘Women of child-bearing age,<br />

including post-partum women’ above).<br />

390 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


A number of commercial assays for testing immunity to rubella are available.<br />

<strong>The</strong>se vary according to the method used to determine the positive cut-off<br />

value (the WHO cut-off is 10 IU/mL, but, at present, there is no recommended<br />

<strong>Australian</strong> minimal level). Available data support the presumption that an<br />

antibody level found by use of a licensed assay to be above the standard positive<br />

cut-off for that assay can be considered evidence of past exposure to rubella<br />

virus. 7 Rubella vaccine induces immune responses that are similar in quality, but<br />

lesser in quantity, than those after natural disease. 3 Measurement of antibody by<br />

commercial assays is not a perfect correlate of protection in vaccinated persons. 3<br />

While on the one hand, those with low levels of vaccine-induced antibodies<br />

are often protected, conversely, reinfection may take place in some individuals<br />

with measurable antibodies. If a person is found to be rubella IgG seronegative,<br />

vaccination should be provided according to the recommendations above.<br />

Interpretation of the results of serological testing may be enhanced by discussion<br />

with the laboratory that performed the test, ensuring that relevant clinical<br />

information is provided. In addition, expert consultation and referral of sera to<br />

a reference laboratory are recommended if there is difficulty interpreting results,<br />

particularly for women of child-bearing age (see ‘Women of child-bearing age,<br />

including post-partum women’ above).<br />

All women of child-bearing age should be advised by a medical practitioner<br />

of the result of their antibody test, as it is a clinically significant test. 39 Women<br />

should be screened for rubella antibodies shortly before every pregnancy, or<br />

early in the pregnancy, or if pregnancy is contemplated, irrespective of a previous<br />

positive rubella antibody result. 3,14 Very occasionally, errors may result in patients<br />

who are seronegative being reported as seropositive. Specimens from pregnant<br />

women are required to be stored until the completion of the pregnancy for<br />

parallel serological testing if required. 40<br />

4.18.8 Pregnancy and breastfeeding<br />

Rubella-containing vaccines are contraindicated in pregnant women<br />

(see 4.18.9 Contraindications below). Pregnancy should be avoided for 28 days<br />

after vaccination. 41<br />

MMR vaccines can be given to breastfeeding women. <strong>The</strong> rubella vaccine<br />

virus may be secreted in human breast milk, and rare cases of transmission of<br />

vaccine virus through breast milk have been reported. However, symptoms<br />

in the newborn have been absent or mild. 42-44 Post-partum vaccination of<br />

women without evidence of rubella immunity need not be delayed because of<br />

breastfeeding.<br />

MMRV vaccines are not recommended for use in persons aged ≥14 years.<br />

<strong>The</strong>re is no risk to pregnant women from contact with recently vaccinated<br />

persons. <strong>The</strong> vaccine virus is not transmitted from vaccinated persons to<br />

susceptible contacts. 1<br />

PART 4 VACCINE-PREVENTABLE DISEASES 391<br />

4.18 RUBELLA


Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.18.9 Contraindications<br />

Anaphylaxis to vaccine components<br />

MMR and MMRV vaccines are contraindicated in persons who have had:<br />

• anaphylaxis following a previous dose of any MMR-containing vaccine<br />

• anaphylaxis following any vaccine component.<br />

Persons who are immunocompromised<br />

Measles-, mumps-, and rubella-containing vaccines contain live<br />

attenuated vaccine viruses and are contraindicated in persons who are<br />

immunocompromised. Thus, MMR-containing vaccines are contraindicated in<br />

the following groups:<br />

• Persons immunocompromised due to HIV/AIDS. MMR vaccination of<br />

asymptomatic HIV-infected persons >12 months of age with an age-specific<br />

CD4 + count of ≥15% may be considered45-48 (see ‘HIV-infected persons’<br />

in 3.3.3 Vaccination of immunocompromised persons). Since studies have not<br />

been performed using combination MMRV vaccines in asymptomatic HIVinfected<br />

persons or persons with an age-specific CD4 + count of ≥15%, it is<br />

recommended that only MMR vaccine (and monovalent VV, see 4.22 Varicella)<br />

be considered for use in this setting. 47,49-51<br />

• Persons with other medical conditions associated with significant<br />

immunocompromise (see 3.3.3 Vaccination of immunocompromised persons)<br />

• Persons receiving high-dose systemic immunosuppressive therapy,<br />

such as chemotherapy, radiation therapy or oral corticosteroids. MMRcontaining<br />

vaccines are contraindicated in persons taking high-dose oral<br />

corticosteroids for more than 1 week (in children equivalent to >2 mg/kg<br />

per day prednisolone, and in adults >60 mg per day) (see 3.3.3 Vaccination<br />

of immunocompromised persons). Those who have been receiving highdose<br />

systemic steroids for more than 1 week may be vaccinated with live<br />

attenuated vaccines after corticosteroid therapy has been discontinued for at<br />

least 1 month52 (see 3.3.3 Vaccination of immunocompromised persons).<br />

See also 3.3 Groups with special vaccination requirements and 4.22 Varicella for more<br />

information.<br />

Pregnant women<br />

See also 4.18.8 Pregnancy and breastfeeding above.<br />

Rubella-containing vaccines are contraindicated in pregnant women.<br />

This is due to the theoretical risk of transmission of the rubella component of the<br />

vaccine to a susceptible fetus. However, no evidence of vaccine-induced CRS has<br />

been reported. 1 Active surveillance in the United States, the United Kingdom and<br />

392 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Germany indicates that no case of vaccine-induced congenital rubella syndrome<br />

occurred among more than 500 women inadvertently vaccinated with rubella<br />

vaccine during pregnancy, whose pregnancies continued. 53 In an Iranian study<br />

performed after mass vaccination with a measles–rubella vaccine, 117 susceptible<br />

women were inadvertently vaccinated while pregnant or became pregnant<br />

within 3 months after vaccination. <strong>The</strong>re were no CRS-related abnormalities<br />

among the infants born to these women. 54 Based on this evidence, the vaccine<br />

cannot be considered to be teratogenic, and termination of pregnancy following<br />

inadvertent vaccination is not indicated. 1,8 (See also 3.3.2 Vaccination of women who<br />

are planning pregnancy, pregnant or breastfeeding, and preterm infants.)<br />

4.18.10 Precautions<br />

For additional precautions related to MMR and MMRV vaccines, see 4.9 Measles<br />

and 4.22 Varicella.<br />

Vaccination with other live attenuated parenteral vaccines<br />

If MMR or MMRV vaccine is not given simultaneously with other live attenuated<br />

parenteral vaccines (e.g. varicella, BCG, yellow fever), the vaccines should be<br />

given at least 4 weeks apart.<br />

Vaccination after immunoglobulin or blood product administration<br />

Administration of a MMR or MMRV vaccine should be delayed after<br />

administration of immunoglobulin-containing products. After receipt of<br />

immunoglobulin-containing blood products, the expected immune response<br />

to measles, mumps, rubella and varicella vaccination may be impaired. 7,55,56<br />

MMR-containing vaccines should not be given for between 3 and 11 months<br />

following the administration of immunoglobulin-containing blood products.<br />

<strong>The</strong> interval between receipt of the blood product and vaccination depends on<br />

the amount of immunoglobulin in each product, and is indicated in 3.3 Groups<br />

with special vaccination requirements, Table 3.3.6 Recommended intervals between<br />

either immunoglobulins or blood products and MMR, MMRV or varicella vaccination. 55<br />

For further information, see 3.3.4 Vaccination of recent recipients of normal human<br />

immunoglobulin and other blood products.<br />

Recent blood transfusion with washed red blood cells is not a contraindication to<br />

MMR or MMRV vaccines.<br />

MMR vaccine may be administered concomitantly with, or at any time in<br />

relation to, anti-D immunoglobulin, but at a separate injection site. Anti-D<br />

immunoglobulin does not interfere with the antibody response to vaccine. 1,3,8<br />

Immunoglobulin or blood product administration after vaccination<br />

Immunoglobulin-containing products should not be administered for 3 weeks<br />

following vaccination with rubella-containing vaccines, unless the benefits<br />

exceed those of vaccination. If immunoglobulin-containing products are<br />

administered within this interval, the vaccinated person should either be<br />

PART 4 VACCINE-PREVENTABLE DISEASES 393<br />

4.18 RUBELLA


evaccinated later at the appropriate time following the product (as indicated in<br />

Table 3.3.6), or be tested for immunity 6 months later and then revaccinated if<br />

seronegative.<br />

Rh (D) immunoglobulin (anti-D) may be given at the same time in different sites<br />

with separate syringes or at any time in relation to MMR vaccine, as it does not<br />

interfere with the antibody response to the vaccine.<br />

4.18.11 Adverse events<br />

Adverse events following administration of MMR-containing vaccines are<br />

generally mild and well tolerated. 3 Adverse events are much less common after<br />

the 2nd dose of MMR or MMRV vaccine than after the 1st dose.<br />

Mild adverse events such as fever, sore throat, lymphadenopathy, rash, arthralgia<br />

and arthritis may occur following MMR vaccination. 1,7 Symptoms most often<br />

begin 1 to 3 weeks after vaccination and are usually transient.<br />

Thrombocytopenia (usually self-limiting) has been very rarely associated with<br />

the rubella or measles component of MMR vaccine, occurring in 3 to 5 per 100 000<br />

doses of MMR vaccine administered. 7,57-59 This is considerably less frequent than<br />

after natural measles, mumps and rubella infections. 59<br />

For further information on adverse events related to MMR and MMRV vaccines,<br />

see 4.9 Measles and 4.22 Varicella.<br />

4.18.12 Public health management of rubella<br />

Rubella is a notifiable disease in all states and territories in Australia.<br />

Further instructions about the public health management of rubella, including<br />

management of cases of rubella and their contacts, should be obtained from state/<br />

territory public health authorities (see Appendix 1 Contact details for <strong>Australian</strong>, state<br />

and territory government health authorities and communicable disease control).<br />

Rubella-containing vaccine does not provide protection if given after exposure to<br />

rubella. 7 However, if the exposure did not result in infection, the vaccine would<br />

induce protection against subsequent infection. Normal human immunoglobulin<br />

(NHIG) has been shown not to be of value in post-exposure prophylaxis for<br />

rubella. 7 However, NHIG may be recommended in certain circumstances (see ‘Use<br />

of normal human immunoglobulin in pregnant women exposed to rubella’ below).<br />

Suspected rubella contacts<br />

All contacts of persons with suspected rubella infection should be identified,<br />

especially those who are pregnant (see ‘Pregnant women with suspected rubella<br />

or exposure to rubella’ below).<br />

Contacts >12 months of age without adequate proof of immunity should receive<br />

1 dose of MMR vaccine (or MMRV vaccine, if appropriate). This will not prevent<br />

rubella disease if already exposed. If vaccination is refused, the contact should<br />

avoid further contact with cases until at least 4 days after onset of the rash in the<br />

394 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


case. Seronegative women of child-bearing age should be vaccinated and tested<br />

for seroconversion 6 to 8 weeks after vaccination (see 4.18.7 Recommendations<br />

above).<br />

Exposed healthcare workers without adequate proof of immunity should be<br />

excluded from work for 21 days from exposure or for at least 4 days after the<br />

onset of rash. 60<br />

Testing for rubella infection<br />

All cases of suspected rubella infection should be laboratory tested and false<br />

positive results excluded (see ‘Serological testing for immunity to rubella’ in<br />

4.18.7 Recommendations above).<br />

Acute rubella infection is indicated by the presence of rubella IgM or a 4-fold or<br />

greater increase in rubella IgG. Rubella IgM may not appear until a week after<br />

clinical symptoms. Sera for testing should be taken 7 to 10 days after onset of<br />

illness and repeated 2 to 3 weeks later. <strong>The</strong> most recent date of potential exposure<br />

should be obtained, if possible, to calculate the potential incubation period. As<br />

some patients may have more than one exposure to a person with a rubella-like<br />

illness, and because exposure may occur over a prolonged period, it is important<br />

to ascertain the dates of the first and last exposures. 60 Testing for infection can<br />

also be done, particularly early in the course of a clinical illness, using virusdetection<br />

methods, such as nucleic acid amplification testing (PCR). 60<br />

Infected persons should be excluded from school/work/institution and should<br />

avoid contact with women of child-bearing age for at least 4 days after the onset<br />

of the rash. 60<br />

Pregnant women with suspected rubella or exposure to rubella<br />

All pregnant women with suspected rubella or exposure to rubella should<br />

be serologically tested (for IgM and IgG), irrespective of a history of prior<br />

vaccination, clinical rubella or a previous positive rubella antibody result (for<br />

more details, see ‘Testing for rubella infection’ above). Testing is essential because<br />

of the serious consequences of the infection, the rash of rubella is not diagnostic,<br />

asymptomatic infection can occur, and the diagnosis requires confirmation by<br />

laboratory tests. 3,7,8 In addition, infection has been reported in women who have<br />

previous evidence of antibody. 7<br />

Serologic specimens should include information regarding the date of the<br />

last menstrual period and the date of presumed exposure (or date of onset of<br />

symptoms). 60 If the woman has an antibody titre below the protective level, or a<br />

low level of antibodies and remains asymptomatic, a second specimen should be<br />

collected 28 days after the exposure (or onset of symptoms) and tested in parallel<br />

with the first. Alternatively, if the woman develops symptoms/signs of rubella<br />

infection, a second serum specimen should be tested as soon as possible. A third<br />

blood specimen may be required in some circumstances. 8 Testing for infection<br />

PART 4 VACCINE-PREVENTABLE DISEASES 395<br />

4.18 RUBELLA


can also be done, particularly early in the course of a clinical illness, using virusdetection<br />

methods, such as nucleic acid amplification testing (PCR). 60<br />

Pregnant women should be counselled to restrict contact with persons with<br />

confirmed, probable or suspected rubella for 6 weeks (2 incubation periods). 60<br />

Counselling of pregnant women with confirmed rubella regarding the risk to the<br />

fetus should be given in conjunction with the woman’s obstetric service.<br />

Use of normal human immunoglobulin in pregnant women exposed to rubella<br />

Post-exposure prophylaxis with normal human immunoglobulin (NHIG) does<br />

not prevent infection in non-immune contacts and is, therefore, of little value<br />

for protection of susceptible contacts exposed to rubella. 7 However, it may<br />

prolong the incubation period. If given to non-immune pregnant contacts, this<br />

may marginally reduce the risk to the fetus. It may also reduce the likelihood of<br />

clinical symptoms in the mother. In such cases, IM administration of 20 mL of<br />

NHIG within 72 hours of rubella exposure might reduce, but will not eliminate,<br />

the risk for rubella. 60 Serological follow-up of recipients is essential, and should<br />

continue for up to 2 months.<br />

<strong>The</strong>re is some evidence to suggest that, in outbreak situations, pre-exposure<br />

NHIG may be effective in preventing infection in women who are likely to be<br />

pregnant, and its use may be indicated for such women with low antibody titres<br />

in high-risk occupations. 61<br />

4.18.13 Variations from product information<br />

<strong>The</strong> product information for MMR and MMRV vaccines recommends that<br />

women of child-bearing age should be advised not to become pregnant for<br />

3 months after vaccination. <strong>The</strong> ATAGI instead recommends avoiding<br />

pregnancy for 28 days after vaccination. 41<br />

For further information on MMR and MMRV vaccines, see 4.9 Measles and<br />

4.22 Varicella.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

396 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.19 TETANUS<br />

4.19.1 Bacteriology<br />

Tetanus is caused by Clostridium tetani, a motile, non-capsulated, Gram-positive<br />

rod that forms endospores. Spores of the bacillus are found in manured soil and<br />

can enter wounds. Once in a wound site, the bacillus can grow anaerobically.<br />

C. tetani produces a potent protein toxin, which has two components,<br />

tetanospasmin (a neurotoxin) and tetanolysin (a haemolysin).<br />

4.19.2 Clinical features<br />

Tetanus is an acute, often fatal, disease caused by the toxin produced by<br />

C. tetani. <strong>The</strong> neurotoxin acts on the central nervous system to cause muscle<br />

rigidity with painful spasms. <strong>The</strong> disease usually occurs after an incubation<br />

period of 3 to 21 days (range 1 day to several months), with a median time<br />

of onset after injury of 10 days. Generally, a shorter incubation period is<br />

associated with a more heavily contaminated wound, more severe disease<br />

and a worse prognosis. Generalised tetanus, the most common form of the<br />

disease, is characterised by increased muscle tone and generalised spasms.<br />

Early symptoms and signs include increased tone in the masseter muscles<br />

(trismus, or lockjaw), dysphagia, and stiffness or pain in the neck, shoulder<br />

and back muscles. Some patients develop paroxysmal, violent, painful,<br />

generalised muscle spasms. A constant threat during generalised spasms is<br />

reduced ventilation, apnoea or laryngospasm. <strong>The</strong> patient may be febrile,<br />

although many have no fever; mental state is unimpaired. Sudden cardiac<br />

arrest sometimes occurs, but its basis is unknown. Other complications include<br />

pneumonia, fractures, muscle rupture, deep vein thrombophlebitis, pulmonary<br />

emboli, decubitus ulcers and rhabdomyolysis. Death results from respiratory<br />

failure, hypertension, hypotension or cardiac arrhythmia.<br />

Tetanus is uncommon in people who have received 4 or more doses of a tetanuscontaining<br />

vaccine and in those who received their last dose within 10 years. 1,2<br />

However, cases have been reported3,4 and clinicians should consider tetanus<br />

when there are appropriate symptoms and signs, irrespective of the person’s<br />

vaccination status. A high level of diagnostic awareness of tetanus is particularly<br />

important in the elderly in industrialised countries, including Australia, as most<br />

deaths occur in people over 70 years of age, especially women, and may be<br />

associated with apparently minor injury. 1,2,5<br />

Neonatal tetanus is usually associated with generalised symptoms, and fatal<br />

if left untreated. It usually occurs following contamination of the umbilical<br />

cord stump. Neonatal tetanus was effectively eliminated in Australia and other<br />

developed countries over a century ago. Introduction of maternal immunisation<br />

during pregnancy with tetanus toxoid has seen neonatal tetanus almost<br />

eliminated in developing countries. 6<br />

PART 4 VACCINE-PREVENTABLE DISEASES 397<br />

4.19 TETANUS


4.19.3 Epidemiology<br />

In Australia, tetanus is rare, occurring primarily in older adults who have<br />

never been vaccinated or who were vaccinated in the remote past. <strong>The</strong>re<br />

were 24 notified cases of tetanus during 2001–2007, but 156 hospitalisations<br />

(July 2000–June 2007) where tetanus was coded as the principal diagnosis. 5,7,8<br />

This discrepancy suggests under-notification. During 2001–2006, there were<br />

3 deaths recorded from tetanus. 5,7,8 <strong>The</strong> case-fatality rate in Australia is about<br />

2%. Effective protection against tetanus can be provided only by active<br />

immunisation. This is because the amount of tetanus toxin required to produce<br />

clinical symptoms is too small to induce a protective antibody response; second<br />

cases of tetanus in unimmunised persons have been recorded. Tetanus vaccine<br />

was introduced progressively into the childhood vaccination schedule after<br />

World War II. <strong>The</strong> effectiveness of the vaccine was demonstrated in that war; all<br />

<strong>Australian</strong> servicemen were vaccinated against tetanus and none contracted the<br />

disease. As tetanus can follow apparently trivial, even unnoticed wounds, active<br />

immunisation is the only certain protection. 1 A completed course of vaccination<br />

provides protection for many years.<br />

4.19.4 Vaccines<br />

Tetanus toxoid is available in Australia only in combination with diphtheria<br />

and other antigens.<br />

<strong>The</strong> acronym DTPa, using capital letters, signifies child formulations of<br />

diphtheria, tetanus and acellular pertussis-containing vaccines. <strong>The</strong> acronym<br />

dTpa is used for formulations that contain substantially lesser amounts<br />

of diphtheria toxoid and pertussis antigens than child (DTPa-containing)<br />

formulations; dTpa vaccines are usually used in adolescents and adults.<br />

Tetanus vaccination stimulates the production of antitoxin. Hence, vaccination<br />

does not prevent growth of C. tetani in contaminated wounds, but protects<br />

against the toxin produced by the organism. <strong>The</strong> immunogen is prepared by<br />

treating a cell-free preparation of toxin with formaldehyde, thereby converting<br />

it into the innocuous tetanus toxoid. Tetanus toxoid is usually adsorbed onto an<br />

adjuvant, either aluminium phosphate or aluminium hydroxide, to increase its<br />

immunogenicity. Antigens from Bordetella pertussis, in combination vaccines, also<br />

act as an effective adjuvant.<br />

Complete immunisation (a 3-dose primary schedule and 2 booster doses) induces<br />

protective levels of antitoxin throughout childhood and into adulthood but, by<br />

middle age, about 50% of vaccinated persons have low or undetectable levels. 9-11<br />

A single dose of tetanus toxoid produces a rapid anamnestic response in such<br />

persons. 12-15<br />

398 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Formulations for children aged


• Tripacel – Sanofi Pasteur Pty Ltd (DTPa; diphtheria-tetanus-acellular<br />

pertussis). Each 0.5 mL monodose vial contains ≥30 IU diphtheria<br />

toxoid, ≥40 IU tetanus toxoid, 10 µg PT, 5 µg FHA, 3 µg PRN, 5 µg<br />

FIM 2+3; 1.5 mg aluminium phosphate; 3.4 mg phenoxyethanol.<br />

Reduced antigen formulations for adults, adolescents and children aged<br />

≥10 years<br />

• ADT Booster – CSL Limited/Statens Serum Institut (dT; diphtheriatetanus).<br />

Each 0.5 mL monodose vial or pre-filled syringe contains<br />

≥2 IU diphtheria toxoid and ≥20 IU tetanus toxoid, adsorbed onto<br />

0.5 mg aluminium as aluminium hydroxide.<br />

• Adacel – Sanofi Pasteur Pty Ltd (dTpa; diphtheria-tetanus-acellular<br />

pertussis). Each 0.5 mL monodose vial contains ≥2 IU diphtheria<br />

toxoid, ≥20 IU tetanus toxoid, 2.5 µg PT, 5 µg FHA, 3 µg PRN,<br />

5 µg FIM 2+3; 0.33 mg aluminium as aluminium phosphate;<br />

phenoxyethanol as preservative; traces of formaldehyde and<br />

glutaraldehyde.<br />

• Adacel Polio – Sanofi Pasteur Pty Ltd (dTpa-IPV; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus). Each 0.5 mL monodose vial<br />

or pre-filled syringe contains ≥2 IU diphtheria toxoid, ≥20 IU tetanus<br />

toxoid, 2.5 µg PT, 5 µg FHA, 3 µg PRN, 5 µg FIM 2+3, 40 D-antigen<br />

units inactivated poliovirus type 1 (Mahoney), 8 D-antigen units type<br />

2 (MEF-1) and 32 D-antigen units type 3 (Saukett); 0.33 mg aluminium<br />

as aluminium phosphate; phenoxyethanol as preservative; traces of<br />

formaldehyde, glutaraldehyde, polysorbate 80, polymyxin, neomycin<br />

and streptomycin.<br />

• Boostrix – GlaxoSmithKline (dTpa; diphtheria-tetanus-acellular<br />

pertussis). Each 0.5 mL monodose vial or pre-filled syringe contains<br />

≥2 IU diphtheria toxoid, ≥20 IU tetanus toxoid, 8 µg PT, 8 µg FHA,<br />

2.5 µg PRN, adsorbed onto 0.5 mg aluminium as aluminium<br />

hydroxide/phosphate; traces of formaldehyde, polysorbate 80 and<br />

glycine.<br />

• Boostrix-IPV – GlaxoSmithKline (dTpa-IPV; diphtheria-tetanusacellular<br />

pertussis-inactivated poliovirus). Each 0.5 mL pre-filled<br />

syringe contains ≥2 IU diphtheria toxoid, ≥20 IU tetanus toxoid, 8 µg<br />

PT, 8 µg FHA, 2.5 µg PRN, 40 D-antigen units inactivated poliovirus<br />

type 1 (Mahoney), 8 D-antigen units type 2 (MEF-1) and 32 D-antigen<br />

units type 3 (Saukett), adsorbed onto 0.5 mg aluminium as aluminium<br />

hydroxide/phosphate; traces of formaldehyde, polysorbate 80,<br />

polymyxin and neomycin.<br />

400 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.19.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 16 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

Infanrix hexa must be reconstituted by adding the entire contents of the syringe<br />

to the vial and shaking until the pellet is completely dissolved. Reconstituted<br />

vaccine should be used as soon as practicable. If storage is necessary, hold at<br />

room temperature for not more than 8 hours.<br />

4.19.6 Dosage and administration<br />

<strong>The</strong> dose of all tetanus-containing vaccines is 0.5 mL, to be given by IM injection.<br />

Do not mix DTPa- or dTpa-containing vaccines or dT vaccine with any other<br />

vaccine in the same syringe, unless specifically registered for use in this way.<br />

4.19.7 Recommendations<br />

Infants and children<br />

Tetanus toxoid is given in combination with diphtheria toxoid and acellular<br />

pertussis as DTPa vaccine. <strong>The</strong> recommended 3-dose primary schedule is at 2,<br />

4 and 6 months of age. <strong>The</strong> 1st dose can be given as early as 6 weeks of age, due<br />

to the high morbidity and occasional mortality associated with pertussis in very<br />

young infants. If the 1st dose is given at 6 weeks of age, the next scheduled doses<br />

should still be given at 4 months and 6 months of age (see 4.12 Pertussis).<br />

A booster dose of tetanus-containing vaccine, usually provided as DTPa-<br />

IPV, is recommended at 4 years of age, but can be given as early as 3.5 years.<br />

For this booster dose, all brands of DTPa-containing vaccines are considered<br />

interchangeable.<br />

Where required, DTPa-containing vaccines can be given for catch-up for either<br />

the primary doses or booster dose in children aged


Adults<br />

Booster vaccination<br />

All adults who reach the age of 50 years without having received a booster dose<br />

of dT in the previous 10 years should receive a further tetanus booster dose. This<br />

should be given as dTpa, to also provide protection against pertussis (see 4.12<br />

Pertussis). This stimulates further production of circulating tetanus antibodies at<br />

an age when waning of diphtheria and tetanus immunity is commencing in the<br />

<strong>Australian</strong> population. 9 Travellers to countries where health services are difficult<br />

to access should be adequately protected against tetanus before departure. <strong>The</strong>y<br />

should receive a booster dose of dT (or dTpa if not given previously) if more than<br />

10 years have elapsed since the last dose of dT-containing vaccine.<br />

For persons undertaking high-risk travel, consider giving a booster dose of either<br />

dTpa or dT (as appropriate) if more than 5 years have elapsed since the last dose<br />

of a dT-containing vaccine.<br />

Primary vaccination<br />

Persons who have not received any tetanus vaccines are also likely to have<br />

missed diphtheria vaccination. <strong>The</strong>refore, 3 doses of dT should be given at<br />

minimum intervals of 4 weeks, followed by booster doses at 10 and 20 years after<br />

the primary course. One of these 3 doses (preferably the 1st) should be given as<br />

dTpa, to also provide additional protection against pertussis. In the event that<br />

dT vaccine is not available, dTpa can be used for all primary doses. However,<br />

this is not recommended routinely because there are no data on the safety,<br />

immunogenicity or efficacy of dTpa in multiple doses for primary vaccination.<br />

For additional information on adults with no history of a primary course of dT<br />

vaccine requiring catch-up, see 2.1.5 Catch-up.<br />

4.19.8 Pregnancy and breastfeeding<br />

Although dT or dTpa vaccines are not routinely recommended for pregnant<br />

women, they can be given under certain circumstances, such as for management<br />

of a tetanus-prone wound (see 4.19.9 Tetanus-prone wounds below) or to prevent<br />

pertussis in pregnant women and their newborns (see 4.12 Pertussis). 17<br />

dT or dTpa vaccines can be given to breastfeeding women.<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.19.9 Tetanus-prone wounds<br />

<strong>The</strong> definition of a tetanus-prone injury is not straightforward, as tetanus<br />

may occur after apparently trivial injury, such as from a rose thorn, or with no<br />

history of injury. It is for this reason that all wounds other than clean, minor<br />

cuts are considered ‘tetanus-prone’. However, there are certain types of wounds<br />

that are more likely to favour the growth of tetanus organisms. <strong>The</strong>se include<br />

compound fractures, bite wounds, deep penetrating wounds, wounds containing<br />

402 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


foreign bodies (especially wood splinters), wounds complicated by pyogenic<br />

infections, wounds with extensive tissue damage (e.g. contusions or burns) and<br />

any superficial wound obviously contaminated with soil, dust or horse manure<br />

(especially if topical disinfection is delayed more than 4 hours). Reimplantation<br />

of an avulsed tooth is also a tetanus-prone event, as minimal washing and<br />

cleaning of the tooth is conducted to increase the likelihood of successful<br />

reimplantation. Persons who inject drugs are also at risk of tetanus, particularly<br />

if skin ‘popping’ is practised. 18 Appropriate tetanus prophylaxis measures<br />

in wound management, including use of tetanus immunoglobulin (TIG), are<br />

outlined in Table 4.19.1.<br />

Adults who have sustained injuries deemed to be tetanus-prone (all wounds<br />

other than clean minor cuts) should receive a booster dose of dT if more than<br />

5 years have elapsed since the last dose of tetanus-containing vaccine (see<br />

Table 4.19.1). As an alternative to dT vaccine after a tetanus-prone wound,<br />

adults can receive dTpa vaccine (see 4.12 Pertussis) to also provide additional<br />

protection against pertussis. 19 In children


Table 4.19.1: Guide to tetanus prophylaxis in wound management<br />

History<br />

of tetanus<br />

vaccination<br />

Time since<br />

last dose<br />

Type of wound DTPa, DTPacombinations,<br />

dT, dTpa, as<br />

appropriate<br />

404 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Tetanus<br />

immunoglobulin*<br />

(TIG)<br />

≥3 doses 10 years Clean minor<br />

wounds<br />

YES NO<br />

All other wounds † YES NO ‡<br />


studies indicate that the adverse reactions to a single dose of dTpa are similar<br />

in adults and adolescents, whether administered shortly (18 months) or at a<br />

longer interval after a previous dose of a vaccine containing tetanus/diphtheria<br />

toxoids. 26-29 (See also 4.12 Pertussis.)<br />

4.19.12 Adverse events<br />

Mild discomfort or pain at the injection site persisting for up to a few days is<br />

common. Uncommon general adverse events following dT vaccine include<br />

headache, lethargy, malaise, myalgia and fever. Anaphylaxis, urticaria and<br />

peripheral neuropathy occur very rarely. Brachial neuritis (inflammation of a<br />

nerve in the arm, causing weakness or numbness) has been described following<br />

the administration of tetanus toxoid-containing vaccines, with an estimated<br />

excess risk of approximately 0.5–1 in 100 000 doses in adults. 30,31 For specific<br />

adverse events following combination vaccines containing both tetanus and<br />

pertussis antigens, see 4.12 Pertussis.<br />

4.19.13 Public health management of tetanus<br />

Tetanus is a notifiable disease in all states and territories in Australia.<br />

Further instructions about the public health management of tetanus, including<br />

management of cases of tetanus, should be obtained from state/territory public<br />

health authorities (see Appendix 1 Contact details for <strong>Australian</strong>, state and territory<br />

government health authorities and communicable disease control).<br />

• Tetanus Immunoglobulin-VF (human; for intramuscular use) – CSL<br />

Limited. 160 mg/mL immunoglobulin (mainly IgG) prepared from<br />

human plasma containing high levels of antibody to the toxin of<br />

Clostridium tetani. Single vials contain 250 IU of human tetanus<br />

antitoxin, with the actual volume stated on the label on the vial. Also<br />

contains glycine.<br />

For information on the definition and management of tetanus-prone wounds,<br />

see 4.19. 9 Tetanus-prone wounds and Table 4.19.1 above. To access tetanus<br />

immunoglobulin (for intramuscular use for management of tetanus-prone<br />

wounds, or intravenous tetanus immunoglobulin for the treatment of clinical<br />

tetanus), contact the <strong>Australian</strong> Red Cross Blood Service (see Part 5 Passive<br />

immunisation).<br />

4.19.14 Variations from product information<br />

<strong>The</strong> product information for Infanrix hexa states that this vaccine is indicated<br />

for primary immunisation of infants from the age of 6 weeks. <strong>The</strong> ATAGI<br />

recommends that this vaccine may also be used for catch-up of the primary<br />

schedule in children


<strong>The</strong> product information for Infanrix IPV states that this vaccine is indicated<br />

for use in a 3-dose primary schedule for immunisation of infants from the<br />

age of 6 weeks and as a single booster dose for children ≤6 years of age who<br />

have previously been vaccinated against diphtheria, tetanus, pertussis and<br />

poliomyelitis. In addition, the ATAGI recommends that this product may<br />

also be used for catch-up of the primary schedule or as a booster in children<br />


<strong>The</strong> product information for Adacel and Boostrix states that there is no<br />

recommendation regarding the timing and frequency of booster doses against<br />

pertussis in adults; however, the ATAGI recommends that pregnant or postpartum<br />

women can receive a booster dose every 5 years and that other adults in<br />

contact with infants and/or at increased risk from pertussis can receive a booster<br />

dose every 10 years.<br />

<strong>The</strong> product information for Boostrix, Boostrix-IPV and Adacel states that<br />

dTpa-containing vaccine should not be given within 5 years of a tetanus<br />

toxoid-containing vaccine. <strong>The</strong> product information for Adacel Polio states that<br />

dTpa-containing vaccine should not be given within 3 years of a tetanus toxoidcontaining<br />

vaccine. <strong>The</strong> ATAGI recommends instead that, if protection against<br />

pertussis is required, dTpa-containing vaccines can be administered at any time<br />

following receipt of a dT-containing vaccine.<br />

<strong>The</strong> product information for Boostrix, Boostrix-IPV, Infanrix hexa and<br />

Infanrix IPV states that these vaccines are contraindicated in children with<br />

encephalopathy of unknown aetiology or with neurologic complications<br />

occurring within 7 days following a vaccine dose. <strong>The</strong> ATAGI recommends<br />

instead that the only contraindication is a history of anaphylaxis to a previous<br />

dose or to any of the vaccine components.<br />

<strong>The</strong> product information for Adacel Polio states that this vaccine is not indicated<br />

following a tetanus-prone wound. <strong>The</strong> ATAGI recommends instead that Adacel<br />

Polio can be administered following a tetanus-prone wound.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

PART 4 VACCINE-PREVENTABLE DISEASES 407<br />

4.19 TETANUS


4.20 TUBERCULOSIS<br />

4.20.1 Bacteriology<br />

Tuberculosis (TB) is caused by organisms of the Mycobacterium tuberculosis<br />

complex (M.TB complex), which are slow-growing, aerobic, acid-fast bacilli.<br />

<strong>The</strong> M.TB complex consists of Mycobacterium tuberculosis, M. bovis, M. microti,<br />

M. canetti and M. africanum, 1 of which M. tuberculosis is the cause of almost all TB<br />

in Australia. 2<br />

4.20.2 Clinical features<br />

As infection is usually air-borne, lung disease is the most common form of<br />

tuberculosis, accounting for approximately 60% of notified TB cases in Australia. 3<br />

Cough, fever, sweats, weight loss and haemoptysis are common symptoms<br />

of pulmonary TB. TB lymphadenitis is the most common extrapulmonary<br />

manifestation, but the disease can occur in any part of the body. Disseminated<br />

disease (miliary TB) and meningeal TB are more common in very young children,<br />

and are among the most serious manifestations of TB disease. 1<br />

Most persons infected with M. tuberculosis remain asymptomatic, but there is<br />

a 10% lifetime risk of developing clinical illness (although the risk can vary<br />

depending on age and immune status), sometimes many years after the original<br />

infection. Infants, the elderly and persons who are immunocompromised, due to<br />

drugs or disease or as a result of adverse socioenvironmental circumstances (e.g.<br />

malnutrition, alcoholism), are more prone to rapidly progressive or generalised<br />

infection. 1,4<br />

4.20.3 Epidemiology<br />

<strong>The</strong> World Health Organization (WHO) declared tuberculosis a global emergency<br />

in 1993, and recent reports have reaffirmed the threat to human health. 5 It is<br />

estimated that in 2010 there were 8.8 million incident cases of TB globally. <strong>The</strong><br />

majority of these cases (81%) were accounted for by 22 high-burden countries,<br />

which all have estimated TB incidences of greater than 40 per 100 000. 6<br />

Approximately 1200 cases of TB are notified to <strong>Australian</strong> health authorities<br />

each year. <strong>The</strong> annual notification rate for TB has been relatively stable at<br />

approximately 5 to 6 cases per 100 000 population since 1985. 2,3 In the southern<br />

states, rates among Indigenous <strong>Australian</strong>s overall are comparable with rates<br />

among <strong>Australian</strong>-born non-Indigenous <strong>Australian</strong>s; however, in some specific<br />

settings (e.g. in the Northern Territory, Far North Queensland and northern<br />

South Australia) rates are higher in Indigenous <strong>Australian</strong>s3 (see 3.1 Vaccination<br />

for Aboriginal and Torres Strait Islander people). Most TB cases in Australia (over<br />

85%) occur in persons born overseas, particularly those born in Asia, southern<br />

and eastern European countries, Pacific island nations, and north and sub-<br />

Saharan Africa. <strong>The</strong> rate of TB in the overseas-born population has been slowly<br />

increasing over the past decade. 3 <strong>The</strong> rate of multi-drug resistant (MDR) TB in<br />

Australia has been low (less than 2% of notified cases); however, the proportion<br />

408 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


of MDR-TB cases identified has increased in recent years. 2,3,7-9 Tuberculosis in<br />

animals (M. bovis) has been eradicated by screening and culling programs. 10<br />

Patients who are immunocompromised are at high risk of developing active<br />

TB if they are infected with M. tuberculosis. 4,5,11 Screening programs in Australia<br />

concentrate on contacts of notified cases and others at increased risk of TB<br />

infection, including refugees and healthcare workers.<br />

4.20.4 Vaccine<br />

• BCG vaccine – Sanofi Pasteur Pty Ltd (live vaccine prepared from an<br />

attenuated strain of Mycobacterium bovis). 1.5 mg lyophilised powder<br />

in a multi-dose vial with separate diluent. Reconstituted vaccine<br />

contains 8–32 x 10 6 colony forming units per mL and monosodium<br />

glutamate 1.5% w/v. May contain trace amounts of polysorbate 80.<br />

Reconstituted volume provides about 10 adult or 20 infant doses.<br />

BCG (bacille Calmette-Guérin) vaccine is a suspension of a live attenuated<br />

strain of M. bovis. Worldwide, there are many BCG vaccines available, but they<br />

are all derived from the strain propagated by the Institute Pasteur, which was<br />

first tested in humans in 1921. 12 BCG vaccination probably has little effect on<br />

preventing infection per se, or reactivation among those already infected with<br />

TB, so the role of BCG vaccination in preventing overall transmission is probably<br />

limited. 13 However, there is strong evidence that BCG vaccination in infancy<br />

provides greater than 70% protection against severe disseminated forms of TB<br />

disease in young children, including miliary TB and TB meningitis. 14-18 TB can be<br />

difficult to diagnose in young children and progression to disseminated TB can<br />

be rapid; they are therefore the primary target for the use of BCG vaccine. <strong>The</strong><br />

efficacy of BCG vaccine against pulmonary disease in adults is less consistent<br />

and has ranged from no protection to 80% in controlled trials. 19 <strong>The</strong> greatest<br />

protection has been observed among skin test-negative adults in North America<br />

and Europe, and the lowest among skin test-positive persons in tropical<br />

settings. 13 <strong>The</strong> reason for the wide variation in measured effectiveness is not clear,<br />

but has been attributed to variability in study quality, differences in BCG strains,<br />

host factors such as age at vaccination and nutritional status, and differences<br />

in the prevalence of infection with environmental mycobacteria. <strong>The</strong> duration<br />

of protection following BCG vaccination has been difficult to measure because<br />

the interval between infection and disease may extend to decades. Benefit from<br />

infant vaccination has been found in studies with follow-up of up to 12 years, but<br />

protection is commonly thought to decline over 10 to 20 years. 13 <strong>The</strong>re is evidence<br />

that memory responses persist for up to 10 to 50 years. 20-22<br />

PART 4 VACCINE-PREVENTABLE DISEASES 409<br />

4.20 TUBERCULOSIS


BCG vaccination has been shown to offer some protection against Mycobacterium<br />

leprae, the causative agent of leprosy. 23<br />

BCG vaccine is not used in the treatment of tuberculosis disease. BCG may be<br />

used as a therapeutic modality for transitional cell carcinoma of the bladder.<br />

4.20.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 24 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

BCG vaccine must be reconstituted by adding the entire contents of the diluent<br />

container to the vial and shaking until the powder is completely dissolved.<br />

Reconstituted vaccine is very unstable and must be stored at +2°C to +8°C and<br />

used within one working session of 4 to 6 hours.<br />

4.20.6 Dosage and administration<br />

<strong>The</strong> dose of BCG vaccine in newborns and infants


BCG vaccination procedures<br />

BCG vaccination should only be given by medical or nursing staff who are<br />

trained in BCG vaccination procedures.<br />

• Use a short (10 mm) 26–27 gauge needle with a short bevel. <strong>The</strong> risk of<br />

spillage can be minimised by using an insulin syringe to which the needle is<br />

already attached.<br />

• Wear protective eye-wear. <strong>The</strong> person to be vaccinated (and the parent/carer<br />

holding a small child being vaccinated) should also wear protective eye-wear.<br />

Eye splashes may ulcerate; if an eye splash occurs, wash the eye with saline<br />

or water immediately.<br />

• Identify the correct injection site. BCG vaccine should be injected into the<br />

skin over the region of insertion of the deltoid muscle into the humerus. This<br />

is just above the midpoint of the upper arm. This site is recommended to<br />

minimise the risk of keloid formation. By convention, the left upper arm is<br />

used wherever possible to assist those who subsequently look for evidence of<br />

BCG vaccination.<br />

• Stretch the skin between a finger and thumb and insert the bevel into the<br />

dermis, bevel uppermost, to a distance of about 2 mm. <strong>The</strong> bevel should be<br />

visible through the transparent epidermis.<br />

• If the injection is not intradermal, withdraw the needle and try again at a new<br />

site. A truly intradermal injection should raise a blanched bleb of about 7 mm<br />

in diameter with the features of peau d’orange (the appearance of orange<br />

peel). Considerable resistance will be felt as the injection is given. If this<br />

resistance is not felt, the needle may be in the subcutaneous tissues.<br />

Response to BCG vaccination<br />

In response to BCG vaccination, a small red papule forms and eventually<br />

ulcerates, usually within 2 to 3 weeks of vaccination. <strong>The</strong> ulcer heals with<br />

minimal scarring over several weeks. <strong>The</strong>re may be swelling and tenderness in<br />

local lymph nodes. While a local reaction represents a normal response to BCG<br />

vaccination, more extensive local reactions are less common (see 4.20. 11 Adverse<br />

events below). Subjects who are given BCG vaccine despite latent or previous<br />

TB infection are likely to experience an accelerated response characterised by<br />

induration within 24 to 48 hours, pustule formation in 5 to 7 days, and healing<br />

within 10 to 15 days.<br />

Clinical trials have not shown a consistent relationship between the size of<br />

tuberculin reactions after BCG vaccination and the level of protection provided.<br />

TST is not recommended to demonstrate immunity after BCG vaccination. 28,29<br />

PART 4 VACCINE-PREVENTABLE DISEASES 411<br />

4.20 TUBERCULOSIS


4.20.7 Recommendations<br />

BCG is not recommended for routine use in the general population, given the<br />

low incidence of TB in Australia and the variable efficacy reported in adults.<br />

However, some groups are at increased of tuberculosis and BCG vaccination<br />

may be warranted for these persons, based on a risk assessment. BCG should be<br />

specifically considered for the following groups.<br />

Aboriginal and Torres Strait Islander neonates30 In some parts of Australia, the incidence of TB is appreciably higher among<br />

Aboriginal and Torres Strait Islander people than <strong>Australian</strong>-born non-<br />

Indigenous <strong>Australian</strong>s, and BCG is recommended for neonates living in<br />

those regions. State and territory guidelines should be consulted for local<br />

recommendations and the geographic areas where BCG vaccination of<br />

Aboriginal and Torres Strait Islander neonates is conducted (see Appendix 1<br />

Contact details for <strong>Australian</strong>, state and territory government health authorities and<br />

communicable disease control). (See also 3.1 Vaccination for Aboriginal and Torres<br />

Strait Islander people.)<br />

Infants born in Australia to migrant parents<br />

TB is rare in infants and young children born in Australia, but infants born to<br />

parents who have migrated from countries with a high TB incidence (i.e. >40 cases<br />

per 100 000 population per year – see 4.20.3 Epidemiology above) may be at higher<br />

risk of TB exposure in their early life. 31 BCG vaccination of these infants is not<br />

routinely recommended because of the uncertainty of the risks and benefits.<br />

Children who will be travelling to high TB incidence settings<br />

<strong>The</strong> risk of TB disease in children travelling to countries with a high TB incidence<br />

(i.e. >40 cases per 100 000 population per year – see 4.20.3 Epidemiology above)<br />

depends on the age of the child, the duration of stay, and the TB incidence at the<br />

destination. Country-specific incidence data are available from the World Health<br />

Organization. 32<br />

<strong>The</strong> need for BCG vaccination should be assessed for young children,<br />

particularly those aged


Occupational groups<br />

<strong>The</strong>re is some evidence that specific occupational groups are at increased risk of<br />

TB, including embalmers, and healthcare workers likely to encounter patients<br />

with TB (e.g. chest clinic staff) or those involved in conducting autopsies. Due<br />

to the limited evidence of benefit of BCG vaccination in adults and interference<br />

of vaccination with interpretation of TST, routine BCG vaccination of persons<br />

within these occupations is not recommended. In occupational settings, TB<br />

prevention and control should be focused around infection control measures,<br />

employment-based TST screening and therapy for latent TB infection. However,<br />

BCG vaccination should be considered for TST-negative healthcare workers who<br />

are at high risk of exposure to drug-resistant TB, due to the difficulty in treating<br />

drug-resistant infection.<br />

4.20.8 Pregnancy and breastfeeding<br />

BCG vaccine is contraindicated in pregnant women.<br />

BCG vaccine can be given to breastfeeding women.<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.20.9 Contraindications<br />

BCG is a live vaccine and its use is contraindicated in the following groups:<br />

• Persons with known or suspected HIV infection, 33 even if asymptomatic<br />

or with normal immune function, because of the risk of disseminated BCG<br />

infection. 34,35<br />

• Persons treated with corticosteroids or other immunosuppressive therapy,<br />

including monoclonal antibodies against tumour necrosis factor-alpha (TNFalpha)<br />

(e.g. infliximab, etanercept, adalimulab). Infants born to mothers<br />

treated with bDMARDS (e.g. TNF-alpha blocking monoclonal antibodies)<br />

in the third trimester of pregnancy frequently have detectable antibodies for<br />

several months and they should not be vaccinated36-38 (see also 3.3 Groups with<br />

special vaccination requirements).<br />

• Persons with congenital cellular immunodeficiencies, including specific<br />

deficiencies of the interferon-gamma pathway.<br />

• Persons with malignancies involving bone marrow or lymphoid systems (see<br />

also 3.3 Groups with special vaccination requirements).<br />

• Persons with any serious underlying illness, including severe malnutrition.<br />

• Pregnant women (BCG vaccine has not been shown to cause fetal damage,<br />

but use of live vaccines in pregnancy is not recommended).<br />

• Persons who have previously had TB or a large (≥5 mm) reaction to a<br />

tuberculin skin test.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 413<br />

4.20 TUBERCULOSIS


4.20.10 Precautions<br />

For those who would otherwise be candidates for BCG, vaccination should be<br />

deferred in the following groups:<br />

• Neonates who are medically unstable, until the neonate is in good medical<br />

condition and ready for discharge from hospital.<br />

• Infants born to mothers who are suspected or known to be HIV-positive,<br />

until HIV infection of the infant can be confidently excluded.<br />

• Persons with generalised septic skin disease and skin conditions such as<br />

eczema, dermatitis and psoriasis.<br />

• Persons being treated for latent TB infection, as the therapy is likely to<br />

inactivate the BCG vaccine.<br />

• Persons who have recently received another parenteral live vaccine (e.g.<br />

MMR, MMRV, varicella, zoster and yellow fever vaccines), until 4 weeks have<br />

elapsed, unless these vaccines are given concurrently with the BCG vaccine.<br />

<strong>The</strong>re are no restrictions on the timing of BCG vaccine in relation to oral live<br />

vaccines.<br />

• Persons with significant febrile illness, until 1 month after recovery.<br />

4.20.11 Adverse events<br />

<strong>The</strong> normal reaction to BCG vaccination has been described above (see<br />

4.20.6 Dosage and administration). About 5% of vaccinated persons experience<br />

adverse events. Injection site abscesses occur in 2.5% of vaccinated persons<br />

and lymphadenitis in 1%, while up to 1% require medical attention. 39 Gross<br />

suppurative or generalised complications of BCG vaccination have been treated<br />

with anti-tuberculosis drugs; however, there is no consensus on the management<br />

of these complications and specialist advice should be sought from local state/<br />

territory tuberculosis services. Anaphylactic reactions have also been reported.<br />

Keloid formation can occur, but the risk is minimised if the injection is not given<br />

higher than the level of insertion of the deltoid muscle into the humerus. <strong>The</strong><br />

overall risk of fatal disseminated infection is extremely low (approximately 1 case<br />

per million vaccinated persons). 13<br />

4.20.12 Public health management of tuberculosis<br />

Tuberculosis is a notifiable disease in all states and territories in Australia.<br />

Further instructions about the public health management of tuberculosis,<br />

including management of cases of tuberculosis and their contacts, should<br />

be obtained from state/territory public health authorities (see Appendix 1<br />

Contact details for <strong>Australian</strong>, state and territory government health authorities and<br />

communicable disease control).<br />

414 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.20.13 Variations from product information<br />

Although the product information for BCG vaccine specifies that vaccine must<br />

be used within 8 hours of reconstitution, the National Tuberculosis Advisory<br />

Committee (NTAC) guidelines recommend that any unused vaccine is discarded<br />

after a working period of 4 to 6 hours.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

PART 4 VACCINE-PREVENTABLE DISEASES 415<br />

4.20 TUBERCULOSIS


4.21 TYPHOID<br />

4.21.1 Bacteriology<br />

Typhoid fever is a clinical syndrome caused by a systemic infection with<br />

Salmonella enterica subspecies enterica serovar Typhi (S. Typhi). Paratyphoid fever,<br />

caused by infection with S. enterica serovar Paratyphi A or B, is similar to, and<br />

often indistinguishable from, typhoid fever. 1 <strong>The</strong> two infections are collectively<br />

known as enteric fever, have largely overlapping geographic distributions, and,<br />

although there is no vaccine specifically targeted against paratyphoid fever,<br />

there is evidence to suggest some cross-protection from the oral live attenuated<br />

typhoid vaccine against Paratyphi B. 2-4<br />

4.21.2 Clinical features<br />

Typhoid fever has a usual incubation period of 7 to 14 days (range 3 to 60 days). 5<br />

Although clinical presentations of typhoid fever can be quite variable, a typical<br />

case presents with a low-grade fever, dull frontal headache, malaise, myalgia,<br />

anorexia and a dry cough. 5 <strong>The</strong> fever tends to increase as the disease progresses;<br />

constipation (more typically diarrhoea in young children), abdominal tenderness,<br />

relative bradycardia and splenomegaly are common. Complications occur in 10<br />

to 15% of patients and tend to occur in patients who have been ill for more than<br />

2 weeks. <strong>The</strong> more important complications include gastrointestinal bleeding,<br />

intestinal perforation and typhoid encephalopathy. 5<br />

Relapse occurs in up to 10% of patients, usually 2 to 3 weeks after the initial fever<br />

resolves. Chronic asymptomatic biliary carriage of S. Typhi occurs in up to 5% of<br />

patients with typhoid fever, even after treatment. Chronic carriage is defined by<br />

the continued shedding of the organism for longer than 1 year. Carriers serve as<br />

an important reservoir in endemic areas and are of public health significance (e.g.<br />

if a carrier works in the food industry). 5<br />

4.21.3 Epidemiology<br />

Humans are the sole reservoir of S. Typhi. It is shed in the faeces of those<br />

who are acutely ill and those who are chronic asymptomatic carriers of<br />

the organism; transmission usually occurs via the ingestion of faecally<br />

contaminated food or water.<br />

<strong>The</strong> vast majority of typhoid fever cases occur in less developed countries, where<br />

poor sanitation, poor food hygiene and untreated drinking water all contribute<br />

to endemic disease, with moderate to high incidence and considerable mortality. 6<br />

Geographic regions with high incidence (>100 cases per 100 000 population<br />

per year) include the Indian subcontinent, most Southeast Asian countries<br />

and several South Pacific nations, including Papua New Guinea. Estimates of<br />

incidence from African countries are more limited. In many regions, particularly<br />

the Indian subcontinent, strains partially or completely resistant to many<br />

antibiotics (including ciprofloxacin) are detected with increasing frequency. 7<br />

416 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


In developed countries, typhoid fever is predominantly a travel-related disease,<br />

with a considerably greater risk following travel to the Indian subcontinent<br />

than to other regions. 8,9 Those who travel to endemic regions to visit friends and<br />

relatives (e.g. immigrants who travel to their former homelands) appear to be<br />

at considerably greater risk of acquiring typhoid fever than other travellers. 8-10<br />

<strong>The</strong>re are typically fewer than 150 cases of typhoid fever reported in Australia<br />

each year, with most following travel to regions with endemic disease. 11<br />

4.21.4 Vaccines<br />

Monovalent typhoid vaccines<br />

• Vivotif Oral – CSL Limited/Crucell Switzerland AG (oral live<br />

attenuated typhoid vaccine). Each enteric-coated capsule contains<br />

≥2 x 109 viable organisms of attenuated S. Typhi strain Ty21a; gelatin;<br />

ethylene glycol; sucrose. 3 capsules in a blister pack.<br />

• Typherix – GlaxoSmithKline (purified Vi capsular polysaccharide<br />

vaccine). Each 0.5 mL pre-filled syringe contains 25 µg Vi<br />

polysaccharide of S. Typhi strain Ty2; phenol as preservative;<br />

phosphate buffer.<br />

• Typhim Vi – Sanofi Pasteur Pty Ltd (purified Vi capsular<br />

polysaccharide vaccine). Each 0.5 mL pre-filled syringe contains 25 µg<br />

Vi polysaccharide of S. Typhi strain Ty2; phenol as preservative;<br />

phosphate buffer.<br />

Combination vaccine that contains S. Typhi<br />

• Vivaxim – Sanofi Pasteur Pty Ltd (formaldehyde-inactivated hepatitis<br />

A virus [GBM strain] and typhoid Vi capsular polysaccharide).<br />

Supplied in a dual-chamber syringe which enables the two vaccines<br />

to be mixed just before administration. Each 1.0 mL dose of mixed<br />

vaccine contains 160 ELISA units of inactivated hepatitis A virus<br />

antigens, 25 µg purified typhoid Vi capsular polysaccharide<br />

strain Ty2; 0.3 mg aluminium as aluminium hydroxide; 2.5 μL<br />

phenoxyethanol; 12.5 µg formaldehyde; traces of neomycin, bovine<br />

serum albumin and polysorbate 80.<br />

<strong>The</strong> attenuated non-pathogenic S. Typhi strain Ty21a was derived by chemical<br />

attenuation of a wild-type strain. Attenuated features of Ty21a include the<br />

absence of the enzyme UDP-galactose-4-epimerase and the Vi capsular<br />

polysaccharide antigen (an important virulence determinant of S. Typhi). <strong>The</strong>se<br />

features partially contribute to the non-pathogenicity and, therefore, the safety of<br />

the oral live vaccine. 12<br />

PART 4 VACCINE-PREVENTABLE DISEASES 417<br />

4.21 TYPHOID


<strong>The</strong> oral vaccine Ty21a strain cannot be detected in faeces more than 3 days<br />

after administration of the vaccine. It stimulates serum IgG, vigorous secretory<br />

intestinal IgA and cell-mediated immune responses. 12 Clinical trials, with<br />

different formulations of the vaccine and with a variety of schedules, have<br />

been undertaken in several countries with endemic typhoid fever (Egypt,<br />

Chile, Indonesia). <strong>The</strong>se have documented varying degrees of protection<br />

against the disease. 5,12<br />

Parenteral Vi polysaccharide vaccines are produced by fermentation of the<br />

Ty2 strain, followed by inactivation with formaldehyde, and then extraction<br />

of the polysaccharide from the supernatant using a detergent. 12 <strong>The</strong> vaccines<br />

elicit prompt serum IgG anti-Vi responses in 85 to 95% of adults and children<br />

>2 years of age. <strong>The</strong> vaccines have also been used in clinical trials in endemic<br />

regions (Nepal, South Africa, China), indicating moderate protection<br />

against typhoid fever. 5,12 As with oral typhoid vaccine, herd protection of<br />

unvaccinated persons living in areas with moderate coverage of parenteral<br />

vaccine has been demonstrated. 13,14<br />

Neither the oral nor the parenteral vaccines have been studied in prospective<br />

clinical trials in travellers to endemic regions. Because many travellers do not<br />

have any naturally acquired immunity, the protection conferred through typhoid<br />

vaccination may be less than that documented in the clinical trials mentioned<br />

above. However, there is circumstantial evidence that the vaccines do provide<br />

protection to travellers to endemic regions, 8,9 and that 3-yearly revaccination is<br />

necessary to prolong the protection. 15<br />

4.21.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 16 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

Because the person to be vaccinated will be responsible for looking after<br />

the course of the oral live attenuated vaccine following purchase, details of<br />

how it should be transported (from the pharmacy to home) and stored in the<br />

refrigerator (at home) must be carefully explained.<br />

4.21.6 Dosage and administration<br />

Oral live attenuated vaccine<br />

<strong>The</strong> vaccine is registered for use in persons ≥6 years of age; it is presented in<br />

a pack of 3 capsules. Each dose (a whole capsule) is the same for both adults<br />

and children.<br />

<strong>The</strong> vaccination schedule consists of 1 capsule of vaccine on days 1, 3 and 5,<br />

taken 1 hour before food. <strong>The</strong> capsule must be swallowed whole with water<br />

and must not be chewed, since the organisms can be killed by gastric acid. Do<br />

not give the vaccine concurrently with antibiotics, or other drugs that are active<br />

against Salmonellae. If possible, antibiotics and other relevant drugs should be<br />

delayed for 3 days after the last dose of the vaccine (see 4.21.10 Precautions below).<br />

418 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


A 4th capsule taken on day 7 has been shown in one large clinical trial to<br />

result in a lower incidence of typhoid fever compared with 3 doses. 12,17<br />

However, giving a 4th dose requires partial use of a second pack.<br />

Co-administration with other vaccines<br />

Oral typhoid vaccine can be administered at the same time as any of the live<br />

parenteral vaccines (including yellow fever vaccine or BCG). 12<br />

<strong>The</strong> oral live attenuated typhoid vaccine should be separated from the<br />

administration of inactivated oral cholera vaccine by an interval of at least<br />

8 hours, and separated from the administration of antibiotics by an interval<br />

of at least 3 days (see 4.21.10 Precautions below).<br />

<strong>The</strong> oral live attenuated typhoid vaccine may be given concurrently with<br />

mefloquine or with atovaquone/proguanil combination (Malarone)<br />

(see 4.21.10 Precautions below).<br />

Parenteral Vi polysaccharide vaccines<br />

Both monovalent typhoid vaccines (Typherix and Typhim Vi) are registered<br />

for use in persons ≥2 years of age. <strong>The</strong> dose of both vaccines is 0.5 mL (for both<br />

adults and children), to be given by IM injection.<br />

<strong>The</strong> dose of the combination typhoid Vi polysaccharide/hepatitis A vaccine<br />

(Vivaxim) is 1 mL to be given by IM injection. Vivaxim is registered for use in<br />

persons aged ≥16 years. (See also 4.4 Hepatitis A.)<br />

Co-administration with other vaccines<br />

Parenteral Vi polysaccharide typhoid vaccines can be given with, or at any<br />

time before or after, other travel vaccines, such as oral cholera or yellow fever<br />

vaccines.<br />

4.21.7 Recommendations<br />

It is recommended that travellers be advised about personal hygiene, food<br />

safety and drinking boiled or bottled water only. <strong>The</strong>y should be advised that<br />

raw (or undercooked) shellfish, salads, cold meats, untreated water and ice (in<br />

drinks) are all potentially ‘high-risk’, as are short (day) trips away from higher<br />

quality accommodation venues.<br />

Oral live attenuated vaccine<br />

Children aged


Children aged ≥6 years and adults<br />

Oral typhoid vaccine in either a 3- or 4-dose schedule is recommended for<br />

children aged ≥6 years and adults who are:<br />

• travelling to endemic regions, where food hygiene may be suboptimal and<br />

drinking water may not be adequately treated<br />

• travelling to endemic regions to visit friends and relatives<br />

• military personnel<br />

• laboratory personnel routinely working with S. Typhi.<br />

<strong>The</strong> addition of a 4th oral dose, on day 7, is an option as there is evidence that<br />

4 doses provides greater protection. 12,17<br />

Revaccination of children aged ≥6 years and adults<br />

<strong>The</strong> optimal timing of revaccination against typhoid fever is uncertain and,<br />

therefore, international recommendations vary considerably. 5,7,9,12<br />

Where continued exposure to S. Typhi exists (such as occurs with either<br />

prolonged travel or residence in an endemic region) and the oral live attenuated<br />

vaccine was used initially, a repeat 3-dose or 4-dose course can be given 3 years<br />

after a 3-dose course, or 5 years after a 4-dose course.<br />

Parenteral Vi polysaccharide vaccines<br />

For further recommendations on the use of the combination typhoid Vi<br />

polysaccharide/hepatitis A vaccine see 4.4 Hepatitis A.<br />

Children aged


4.21.8 Pregnancy and breastfeeding<br />

<strong>The</strong> oral live attenuated typhoid vaccine is contraindicated in pregnant women<br />

(see 4.21.9 Contraindications below).<br />

<strong>The</strong> oral live attenuated typhoid vaccine can be given to breastfeeding women.<br />

Parenteral Vi polysaccharide vaccines are not routinely recommended for<br />

pregnant or breastfeeding women, but can be given where vaccination is<br />

considered necessary (see 4.21.7 Recommendations above).<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.21.9 Contraindications<br />

<strong>The</strong> only absolute contraindications to typhoid vaccines are:<br />

• anaphylaxis following a previous dose of any typhoid vaccine<br />

• anaphylaxis following any vaccine component.<br />

Oral live attenuated vaccine<br />

<strong>The</strong> oral live attenuated vaccine should not be administered to:<br />

• children


used, it is recommended that vaccination should be timed so that the last dose of<br />

vaccine is administered at least 3 days before starting antibiotics or antimalarial<br />

prophylaxis.<br />

4.21.11 Adverse events<br />

Typhoid vaccines, both oral and parenteral, are associated with very few adverse<br />

events and, when adverse events do occur, they tend to be mild and transient. 18<br />

Oral live attenuated vaccine<br />

Abdominal discomfort, diarrhoea, nausea, vomiting and rashes have occasionally<br />

been reported.<br />

Parenteral Vi polysaccharide vaccines<br />

Local adverse events such as erythema, swelling and pain at the injection site<br />

occur very commonly in 10 to 20% of vaccine recipients. Systemic adverse events<br />

are common and include fever (3% of recipients), malaise and nausea.<br />

4.21.12 Public health management of typhoid fever<br />

Typhoid fever is a notifiable disease in all states and territories in Australia.<br />

Further instructions about the public health management of typhoid fever,<br />

including management of cases of typhoid fever and their contacts, should<br />

be obtained from state/territory public health authorities (see Appendix 1<br />

Contact details for <strong>Australian</strong>, state and territory government health authorities and<br />

communicable disease control).<br />

4.21.13 Variations from product information<br />

<strong>The</strong> <strong>Australian</strong> product information for Vivotif Oral live attenuated vaccine<br />

does not mention the use of a 4-dose course of the vaccine for either initial or<br />

repeat vaccination, although this vaccine is registered for use in some other<br />

countries (e.g. Canada and the United States) in a 4-dose schedule. <strong>The</strong> ATAGI<br />

recommends that a 4-dose course can be given to provide increased protection<br />

against typhoid fever.<br />

<strong>The</strong> product information for Vivotif Oral live attenuated vaccine does not include<br />

pregnancy among the listed contraindications. <strong>The</strong> ATAGI recommends that<br />

pregnancy is a contraindication to the oral live attenuated typhoid vaccine.<br />

<strong>The</strong> product information for Typhim Vi recommends a booster dose every 2 to<br />

3 years, and the product information for Vivotif Oral live attenuated vaccine<br />

recommends a booster every 3 years. <strong>The</strong> ATAGI also recommends, for those<br />

at continuing risk, revaccination with a dose of parenteral Vi polysaccharide<br />

vaccine every 3 years after a previous dose, or revaccination with a 3- or 4-dose<br />

course of the oral live attenuated vaccine 3 years after a 3-dose course or 5 years<br />

after a 4-dose course.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

422 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.22 VARICELLA<br />

4.22.1 Virology<br />

Varicella-zoster virus (VZV) is a DNA virus within the herpes virus family. 1<br />

Primary infection with VZV causes varicella (chickenpox). Following primary<br />

infection, VZV establishes latency in the dorsal root ganglia. Reactivation of the<br />

latent virus manifests as herpes zoster (shingles) 2 (see 4.24 Zoster).<br />

4.22.2 Clinical features<br />

Varicella is a highly contagious infection spread by respiratory secretions,<br />

including aerosol transmission, or from the vesicle fluid of the skin lesions of<br />

varicella or zoster infection. 1 Varicella is usually a mild disease of childhood.<br />

However, complications occur in approximately 1% of cases. 3 It is more severe<br />

in adults and in persons of any age who are immunocompromised, in whom<br />

complications, disseminated disease and fatal illness can occur. 1<br />

<strong>The</strong> average incubation period is 14 to 16 days (range 10 to 21 days), but may<br />

be longer in persons who are immunocompromised, especially after receipt<br />

of zoster immunoglobulin (ZIG). 2 <strong>The</strong> period of infectivity is from 48 hours<br />

before the onset of rash until crusting of all lesions has occurred. 4 A short<br />

prodromal period of 1 to 2 days may precede the onset of the rash, especially<br />

in adults. 1,2 In otherwise healthy children, skin lesions usually number between<br />

200 and 500. 1,2 Acute varicella may be complicated by secondary bacterial skin<br />

infection, pneumonia, acute cerebellar ataxia (1 in 4000 cases), aseptic meningitis,<br />

transverse myelitis, encephalitis (1 in 100 000 cases) and thrombocytopenia. In<br />

rare cases, it involves the viscera and joints. 1<br />

Congenital varicella syndrome has been reported after varicella infection in<br />

pregnancy and may result in skin scarring, limb defects, ocular anomalies<br />

and neurologic malformations. 1,5 <strong>The</strong>re is a higher risk to the fetus if maternal<br />

infection occurs in the second trimester compared with infection in the first<br />

trimester (1.4% versus 0.55%). 6 Infants with intrauterine exposure also risk<br />

developing herpes zoster in infancy (0.8–1.7%), with the greatest risk following<br />

exposure in the third trimester. 5 Severe neonatal varicella infection can result<br />

from perinatal maternal varicella. 7 <strong>The</strong> onset of varicella in pregnant women<br />

from 5 days before delivery to 2 days after delivery is estimated to result in<br />

severe varicella in 17 to 30% of their newborn infants. 1,7<br />

Reactivation of latent VZV as a result of waning cellular immunity results in<br />

herpes zoster (HZ), a localised vesicular rash. HZ can occur at any age, but<br />

is more common in older adults and persons who are immunocompromised.<br />

Complications may include post-herpetic neuralgia and disseminated zoster with<br />

visceral, central nervous system and pulmonary involvement1 (see 4.24 Zoster).<br />

<strong>The</strong>re is no specific therapy for uncomplicated varicella infection. Antiviral<br />

therapy is used in the treatment of complicated or severe varicella, herpes zoster<br />

disease, and disease in persons who are immunocompromised.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 423<br />

4.22 VARICELLA


4.22.3 Epidemiology<br />

In an unimmunised population in temperate climates, the annual number<br />

of cases of varicella approximates the birth cohort. 8 Tropical regions have<br />

a higher proportion of cases in adults. Approximately 5% of cases are<br />

subclinical. A serosurvey conducted in 1997–1999 found that 83% of the<br />

<strong>Australian</strong> population were seropositive by 10–14 years of age. 9 Prior to the<br />

introduction of a varicella vaccination program in Australia, there were about<br />

240 000 cases, 1500 hospitalisations and an average of 7 to 8 deaths each year<br />

from varicella in Australia. 10-12 <strong>The</strong> highest rates of hospitalisation occur in<br />

children


as a case of wild-type varicella occurring more than 42 days after vaccination.<br />

<strong>The</strong> majority of cases of breakthrough varicella are mild with fewer lesions than<br />

natural infection. However, breakthrough varicella infections can be contagious,<br />

particularly if many lesions are present. 28<br />

Post-marketing studies in the United States have estimated the effectiveness of<br />

1 dose of VV in children to be 80 to 85% against any disease and 95 to 98%<br />

against severe varicella. 28-32 Although earlier data suggested persistence of<br />

immunity in most healthy vaccine recipients, 1 some, but not all, long-term<br />

follow-up studies have shown that rates of vaccine failure increased over time<br />

in 1-dose vaccine recipients. For example, in one study, vaccine failure was<br />

increased 2.6 times in children who received 1 dose of vaccine more than 5 years<br />

previously, compared with those who had received 1 dose of vaccine within<br />

5 years. 33 Follow-up from a randomised controlled trial in children 12 months<br />

to 12 years of age, comparing 1 dose with 2 doses of VV over a 10-year period,<br />

showed significantly higher protection with 2 doses (98.3% versus 94.4%). 34<br />

Based on current evidence, 2 doses of a varicella-containing vaccine in children<br />

from 12 months of age will minimise the risk of breakthrough varicella (see 4.22.7<br />

Recommendations below).<br />

Healthy adolescents (≥14 years of age ) and adults require 2 doses of varicella<br />

vaccine, at least 4 weeks apart, as the response to a single dose of VV decreases<br />

progressively as age increases and is insufficient to provide adequate protection. 35<br />

Combination MMRV vaccines have been shown in clinical trials, conducted<br />

predominantly in children 12 months to 6 years of age, to produce similar rates<br />

of seroconversion to all four vaccine components compared with MMR and<br />

monovalent varicella vaccines administered concomitantly at separate injection<br />

sites. 36-39 In one comparative study assessing seroresponses to a single MMRV<br />

vaccine dose in 12–14-month-old children, the seroresponse rates to measles,<br />

mumps and rubella were similar, but varicella seroresponses were lower in<br />

Priorix-tetra recipients than in ProQuad recipients. 40 However, the clinical<br />

significance of this is not clear, particularly for MMRV given after MMR vaccine.<br />

Monovalent varicella vaccines (VV)<br />

• Varilrix– GlaxoSmithKline (live attenuated Oka strain of varicellazoster<br />

virus). Lyophilised powder in a monodose vial with separate<br />

diluent. Each 0.5 mL reconstituted dose contains ≥103.3 plaqueforming<br />

units (PFU) of attenuated varicella-zoster virus; human<br />

albumin; lactose; neomycin; polyalcohols.<br />

• Varivax Refrigerated – CSL Limited/Merck & Co Inc (live attenuated<br />

Oka/Merck strain of varicella-zoster virus). Lyophilised powder<br />

in a monodose vial with a pre-filled diluent syringe. Each 0.5 mL<br />

PART 4 VACCINE-PREVENTABLE DISEASES 425<br />

4.22 VARICELLA


econstituted dose contains ≥1350 PFU of attenuated varicella-zoster<br />

virus; sucrose; hydrolysed gelatin; urea; monosodium glutamate;<br />

residual components of MRC-5 cells; traces of neomycin and bovine<br />

serum.<br />

Quadrivalent measles-mumps-rubella-varicella (MMRV) vaccines<br />

• Priorix-tetra – GlaxoSmithKline (live attenuated measles virus<br />

[Schwarz strain], mumps virus [RIT 4385 strain, derived from the<br />

Jeryl Lynn strain], rubella virus [Wistar RA 27/3 strain] and varicellazoster<br />

virus [Oka strain]). Lyophilised pellet in a monodose vial with<br />

a pre-filled diluent syringe. Each 0.5 mL reconstituted dose contains<br />

≥103.0 cell culture infectious dose 50% (CCID ) of the Schwarz measles<br />

50<br />

virus, ≥104.4 CCID of the RIT 4385 mumps virus, ≥10 50 3.0 CCID of the<br />

50<br />

Wistar RA 27/3 rubella virus, and ≥103.3 PFU of Oka varicella-zoster<br />

virus; lactose; neomycin; sorbitol; mannitol.<br />

• ProQuad – CSL Limited/Merck & Co Inc (live attenuated measles<br />

virus [Enders’ attenuated Edmonston strain], mumps virus [Jeryl<br />

Lynn B level strain], rubella virus [Wistar RA 27/3 strain] and<br />

varicella-zoster virus [Oka/Merck strain]). Lyophilised powder<br />

in a monodose vial with a pre-filled diluent syringe. Each 0.5 mL<br />

reconstituted dose contains ≥10 3.0 tissue culture infectious dose 50%<br />

(TCID 50 ) of Enders’ attenuated Edmonston measles virus, ≥10 4.3<br />

TCID 50 of the Jeryl Lynn B level mumps virus, ≥10 3.0 TCID 50 of the<br />

Wistar RA 27/3 rubella virus, and ≥10 3.99 PFU of Oka/Merck varicella<br />

virus; sucrose; hydrolysed gelatin; urea; sorbitol; monosodium<br />

L-glutamate; human albumin; neomycin; residual components of<br />

MRC-5 cells; bovine serum albumin.<br />

4.22.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 41 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

Varicella-containing vaccines are less stable than other commonly used live<br />

viral vaccines, and adherence to storage and reconstitution requirements is very<br />

important. All vaccines must be reconstituted by adding the entire contents of the<br />

diluent to the vial containing the pellet, and shaking until the pellet is completely<br />

dissolved. Available monovalent VVs and MMRV vaccines have different<br />

requirements following reconstitution.<br />

Reconstituted Varilrix vaccine should be used as soon as practicable. If storage is<br />

necessary, hold at ambient temperature for not more than 90 minutes, or at +2°C<br />

to +8°C for not more than 8 hours.<br />

426 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Reconstituted Varivax Refrigerated vaccine must be used within 2½ hours.<br />

Reconstituted Priorix-tetra (MMRV) vaccine should be used as soon as practicable.<br />

If storage is necessary, hold at +2°C to +8°C for not more than 8 hours.<br />

Reconstituted ProQuad (MMRV) vaccine must be used within 30 minutes.<br />

4.22.6 Dosage and administration<br />

<strong>The</strong> dose of VV and MMRV vaccines is 0.5 mL, to be given by SC injection.<br />

Priorix-tetra may also be given by IM injection. 42<br />

MMRV vaccines are not recommended for use in persons aged ≥14 years.<br />

<strong>The</strong> minimum interval between doses of varicella-containing vaccine is 4 weeks.<br />

Co-administration with other vaccines<br />

VV and MMRV vaccines can be given at the same time as other live attenuated<br />

parenteral vaccines (e.g. BCG, yellow fever) or other inactivated vaccines<br />

(including DTPa, hepatitis B, Hib, IPV, MenCCV, hepatitis A and pneumococcal<br />

conjugate vaccine), 40 using separate syringes and injection sites. If VV or MMRV<br />

vaccine is not given simultaneously with other live attenuated parenteral<br />

vaccines, they should be given at least 4 weeks apart.<br />

If VV is given at the same time as MMR vaccine, they should be given using<br />

separate syringes and injection sites. MMR vaccine and monovalent VV should<br />

not be mixed together prior to injection.<br />

Interchangeability of varicella-containing vaccines<br />

In general, the two brands of varicella vaccine can be considered interchangeable;<br />

that is, the 2nd varicella dose does not have to be of the same brand as the 1st.<br />

<strong>The</strong> same principle applies to the two available MMRV vaccines, 40 although they<br />

are not routinely recommended in a 2-dose schedule.<br />

4.22.7 Recommendations<br />

Children (aged


outbreak. However, note that MMRV vaccine is not recommended for use as the<br />

1st dose of MMR-containing vaccine in children aged


adequate protection from varicella. 35,43 <strong>The</strong> 2 doses should be administered<br />

at least 4 weeks apart. However, a longer interval between vaccine doses is<br />

acceptable. Lack of immunity to varicella should be based on a negative history<br />

of previous varicella infection and can be supplemented by serological testing for<br />

evidence of past infection (see ‘Serological testing for varicella immunity from<br />

infection and/or vaccination’ below).<br />

MMRV vaccines are not recommended for use in persons ≥14 years of age, due to<br />

a lack of data on safety and immunogenicity/efficacy in this age group. If a dose<br />

of MMRV vaccine is inadvertently given to an older person, this dose does not<br />

need to be repeated.<br />

Serological testing for varicella immunity from infection and/or vaccination<br />

Children who have an uncertain clinical history or no documentation of<br />

age-appropriate varicella vaccination should be considered susceptible and<br />

offered vaccination. Although a reliable history of varicella infection correlates<br />

highly with serological evidence of immunity in young children, 44,45 due to the<br />

decreasing incidence of varicella in Australia and reduced familiarity with the<br />

disease, vaccination should be offered, unless confident clinical diagnosis of<br />

prior natural infection is made. Testing of children to assess serologic status<br />

prior to vaccination is generally not recommended. Provided there are no<br />

contraindications, children can safely receive either VV or MMRV vaccine, even if<br />

prior infection with VZV has occurred (see ‘Children (aged


with VZV. (See also 4.22.12 Public health management of varicella below.) Postexposure<br />

vaccination is generally successful when given within 3 days, and up<br />

to 5 days, after exposure, with earlier administration being preferable. 51-55 MMRV<br />

vaccine can be given to children in this setting, particularly if MMR vaccination is<br />

also indicated (see 4.22.7 Recommendations above).<br />

Household contacts of persons who are immunocompromised<br />

Vaccination of household contacts of persons who are immunocompromised is<br />

strongly recommended. This is based on evidence that transmission of varicella<br />

vaccine virus strain is extremely rare and it is likely to cause only mild disease<br />

(see 4.22.11 Adverse events below). This compares with the relatively high risk<br />

of severe varicella disease from exposure to wild-type varicella-zoster virus<br />

in persons who are immunocompromised. 49,56 If vaccinated persons develop<br />

a rash, they should cover the rash and avoid contact with persons who are<br />

immunocompromised for the duration of the rash. Zoster immunoglobulin<br />

(ZIG) need not be given to an immunocompromised contact of a vaccinated<br />

person with a rash, because the disease associated with this type of transmission<br />

(should it occur) is expected to be mild (see 4.22.12 Public health management of<br />

varicella below).<br />

Healthcare workers, staff working in early childhood education and care, and in<br />

long-term care facilities<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.7 Recommended<br />

vaccinations for persons at increased risk of certain occupationally acquired vaccinepreventable<br />

diseases for more information.<br />

Vaccination against varicella is recommended for all non-immune adults, but<br />

especially for all healthcare workers (HCW), staff working in early childhood<br />

education and care, and staff working in long-term care facilities. Persons in<br />

such occupations who have a negative or uncertain history of varicella infection,<br />

and who do not have documentation of 2 doses of varicella vaccine, should<br />

be vaccinated with 2 doses of varicella vaccine or have serological evidence of<br />

immunity to varicella57 (see ‘Adolescents (aged ≥14 years) and adults’ above).<br />

Testing to check for seroconversion after VV is not recommended (see ‘Serological<br />

testing for varicella immunity from infection and/or vaccination’ above).<br />

However, since varicella vaccination is not 100% effective, HCWs and other<br />

carers should still be advised of the signs and symptoms of infection and how to<br />

manage them appropriately according to local protocols if they develop varicella.<br />

4.22.8 Pregnancy and breastfeeding<br />

Varicella-containing vaccines are contraindicated in pregnant women (see<br />

4.22.9 Contraindications below). Pregnancy should be avoided for 28 days after<br />

vaccination.<br />

Varicella-containing vaccines can be given to breastfeeding women. Most live<br />

vaccines have not been demonstrated to be secreted in breast milk. Women who<br />

430 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


eceived varicella vaccine while breastfeeding showed no evidence of VZV DNA<br />

in breast milk samples, and no effects on breastfed infants have been reported. 58<br />

Post-partum vaccination of women without evidence of varicella immunity need<br />

not be delayed because of breastfeeding.<br />

MMRV vaccines are not recommended for use in persons aged ≥14 years.<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.22.9 Contraindications<br />

Anaphylaxis to vaccine components<br />

Varicella-containing vaccines are contraindicated in persons who have had:<br />

• anaphylaxis following a previous dose of any varicella-containing vaccine<br />

• anaphylaxis following any vaccine component.<br />

Persons who are immunocompromised<br />

Measles-, mumps-, rubella- and varicella-containing vaccines contain live<br />

attenuated vaccine viruses and are contraindicated in persons who are<br />

immunocompromised. Thus, both VV and MMRV vaccines are contraindicated<br />

in the following groups:<br />

• Persons immunocompromised due to HIV/AIDS. Vaccination with live<br />

attenuated vaccines can result in a more extensive vaccine-associated rash<br />

or disseminated infection in persons with AIDS. 59-62 However, varicella<br />

vaccination (with a 2-dose schedule of VV) of asymptomatic HIV-infected<br />

persons >12 months of age with an age-specific CD4 + count of ≥15%<br />

may be considered63-65 (see ‘HIV-infected persons’ in 3.3.3 Vaccination of<br />

immunocompromised persons). Since studies have not been performed using<br />

combination MMRV vaccines in asymptomatic HIV-infected persons<br />

or persons with an age-specific CD4 + count of ≥15%, it is recommended<br />

that only MMR vaccine and monovalent VV be considered for use in this<br />

setting. 60,64,65<br />

• Persons with other medical conditions associated with significant<br />

immunocompromise (see 3.3.3 Vaccination of immunocompromised persons).<br />

• Persons receiving high-dose systemic immunosuppressive therapy, such<br />

as chemotherapy, radiation therapy or oral corticosteroids. Varicellacontaining<br />

vaccines are contraindicated in persons taking high-dose oral<br />

corticosteroids for more than 1 week (in children equivalent to >2 mg/kg<br />

per day prednisolone, and in adults >60 mg per day) (see 3.3.3 Vaccination<br />

of immunocompromised persons). Those who have been receiving highdose<br />

systemic steroids for more than 1 week may be vaccinated with live<br />

attenuated vaccines after corticosteroid therapy has been discontinued for at<br />

least 1 month66 (see 3.3.3 Vaccination of immunocompromised persons).<br />

PART 4 VACCINE-PREVENTABLE DISEASES 431<br />

4.22 VARICELLA


See also 3.3 Groups with special vaccination requirements and 4.9 Measles for more<br />

information.<br />

Pregnant women<br />

See also 4.22.8 Pregnancy and breastfeeding above.<br />

Varicella-containing vaccines are contraindicated in pregnant women.<br />

This is due to the theoretical risk of transmission of the varicella component<br />

of the vaccine to a susceptible fetus. However, no evidence of vaccine-induced<br />

congenital varicella syndrome has been reported. Data from a registry,<br />

established in the United States to monitor the maternal–fetal outcomes of<br />

pregnant women who were inadvertently administered VV either 3 months<br />

before, or at any time during, pregnancy, showed that, among the 587<br />

prospectively enrolled women (including 131 live births to women known to be<br />

varicella-zoster virus-seronegative), there was no evidence of congenital varicella<br />

syndrome. 67 <strong>The</strong> rate of occurrence of congenital anomalies from prospective<br />

reports in the registry was similar to reported rates in the general United States<br />

population (3.2%) and the anomalies showed no specific pattern or target organ.<br />

A non-immune pregnant household contact is not a contraindication to<br />

vaccination with varicella-containing vaccines of a healthy child or adult in the<br />

same household. <strong>The</strong> benefit of reducing the exposure to varicella by vaccinating<br />

healthy contacts of non-immune pregnant women outweighs any theoretical<br />

risks of transmission of vaccine virus to these women.<br />

4.22.10 Precautions<br />

For additional precautions related to MMRV vaccines, see 4.9 Measles.<br />

Vaccination with other live attenuated parenteral vaccines<br />

If a varicella-containing vaccine is not given simultaneously with other live<br />

attenuated parenteral vaccines (e.g. MMR, BCG, yellow fever), the vaccines<br />

should be given at least 4 weeks apart.<br />

Vaccination after immunoglobulin or blood product administration<br />

Administration of MMR or MMRV vaccine should be delayed after<br />

administration of immunoglobulin-containing products. After receipt of<br />

immunoglobulin-containing blood products, the expected immune response<br />

to measles, mumps, rubella and varicella vaccination may be impaired. 68-70<br />

VV or MMRV vaccine should not be given for between 3 and 11 months<br />

following the administration of immunoglobulin-containing products. <strong>The</strong><br />

interval between receipt of the blood product and vaccination depends on the<br />

amount of immunoglobulin in each product, and is indicated in 3.3 Groups with<br />

special vaccination requirements, Table 3.3.6 Recommended intervals between either<br />

immunoglobulins or blood products and MMR, MMRV or varicella vaccination. 69<br />

For further information, see 3.3.4 Vaccination of recent recipients of normal human<br />

432 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


immunoglobulin and other blood products and 4.22.13 Variations from product<br />

information below.<br />

Recent blood transfusion with washed red blood cells is not a contraindication to<br />

VV or MMRV vaccines.<br />

Varicella vaccine may be administered concomitantly with, or at any time in<br />

relation to, anti-D immunoglobulin, but at a separate injection site. Anti-D<br />

immunoglobulin does not interfere with the antibody response to vaccine.<br />

Immunoglobulin or blood product administration after vaccination<br />

Immunoglobulin-containing products should not be administered for 3 weeks<br />

following vaccination with varicella-containing vaccines, unless the benefits<br />

exceed those of vaccination. If immunoglobulin-containing products are<br />

administered within this interval, the vaccinated person should be revaccinated<br />

later at the appropriate time following the product (as indicated in Table 3.3.6<br />

Recommended intervals between either immunoglobulins or blood products and MMR,<br />

MMRV or varicella vaccination).<br />

Rh (D) immunoglobulin (anti-D) may be given at the same time, in different sites<br />

with separate syringes, or at any time in relation to varicella vaccine, as it does<br />

not interfere with the antibody response to the vaccine.<br />

Persons receiving long-term aspirin or salicylate therapy<br />

Persons receiving long-term salicylate therapy (aspirin) should be vaccinated if<br />

indicated, as the benefit is likely to outweigh any possible risk of Reye syndrome<br />

occurring after vaccination. Natural varicella infection and salicylate use has<br />

been associated with an increased risk of developing Reye syndrome. However,<br />

there have been no reports of an association between Reye syndrome and<br />

varicella vaccination (see 4.22.13 Variations from product information below).<br />

4.22.11 Adverse events<br />

If using MMRV vaccine, additional adverse events relating to the measles,<br />

mumps and rubella vaccine components are discussed in 4.9 Measles, 4.11 Mumps<br />

and 4.18 Rubella.<br />

Adverse events following administration of varicella-containing vaccines are<br />

generally mild and well tolerated. 71<br />

Injection site reactions (pain, redness or swelling) are the most common adverse<br />

events reported after varicella vaccination, occurring in 7 to 30% of vaccine<br />

recipients, but are generally well tolerated. 2,71<br />

A maculopapular or papulovesicular rash may develop after varicella<br />

vaccination (usually within 5 to 26 days). A VV-associated rash is likely to<br />

occur in less than 5% of vaccine recipients, and to last for less than 1 week. 72,73<br />

Rashes typically consist of 2 to 5 lesions and may be generalised (3–5%), or also<br />

commonly occur at the injection site (3–5%). 66 VV-associated rash may be atypical<br />

and may not be vesicular. Most varicelliform rashes that occur within the first<br />

PART 4 VACCINE-PREVENTABLE DISEASES 433<br />

4.22 VARICELLA


2 weeks after vaccination are due to wild-type VZV, with median onset 8 days<br />

after vaccination (range 1 to 24 days), while vaccine-strain VZV rashes occur at a<br />

median of 21 days after vaccination (range 5 to 42 days). 74,75<br />

Transmission of vaccine virus to contacts of vaccinated persons is rare. In the<br />

United States, where more than 56 million doses of VV were distributed between<br />

1995 and 2005, there have been only six well-documented cases of transmission<br />

of the vaccine-type virus from five healthy vaccine recipients who had a vaccineassociated<br />

rash. 66,76 Contact cases have been mild. 66,76-78<br />

Fever >39°C has been observed in 15% of healthy children after varicella<br />

vaccination, but this was comparable to that seen in children receiving placebo. 66<br />

In adults and adolescents, fever has been reported in 10% of VV recipients. It is<br />

recommended that parents/carers/vaccine recipients be advised about possible<br />

symptoms, and given advice for reducing fever, including the use of paracetamol<br />

for fever in the period 5 to 12 days after vaccination. Higher rates of fever were<br />

observed in clinical trials of both MMRV vaccines, particularly following dose 1,<br />

when compared with giving MMR vaccine and monovalent VV at the same<br />

time but at separate sites. 36-39 Two post-marketing studies in the United States<br />

identified an approximately 2-fold increased risk of fever and febrile convulsions<br />

in 1st dose recipients of MMRV vaccine, who were predominantly 12–23 months<br />

of age, in the period 7 to 10 days79 (or 5 to 12 days) 80 after vaccination, compared<br />

with recipients of separate MMR and VV vaccines. MMRV vaccination resulted<br />

in 1 additional febrile seizure for every 2300 doses compared to separate MMR<br />

and VV vaccination. 79 An increase in fever or febrile convulsions has not been<br />

identified after the 2nd dose of MMRV vaccine in the United States, although<br />

most 2nd dose recipients were aged 4–6 years, an age at which the incidence of<br />

febrile convulsions is low. 81 <strong>The</strong>se post-marketing studies were in children who<br />

received ProQuad; however, it is anticipated that this side effect profile would be<br />

similar in Priorix-tetra recipients.<br />

A post-marketing study in the United States reported serious adverse events<br />

temporally, but not necessarily causally, linked to varicella vaccination, such<br />

as encephalitis, ataxia, thrombocytopenia and anaphylaxis, were very rare and<br />

occurred in


eported in a 9-year follow-up of 7000 varicella vaccinated children. 27 (See also<br />

4.24 Zoster.)<br />

4.22.12 Public health management of varicella<br />

Varicella is a notifiable disease in most states and territories in Australia.<br />

Further instructions about the public health management of varicella, including<br />

management of cases of varicella and their contacts, should be obtained from<br />

state/territory public health authorities (see Appendix 1 Contact details for<br />

<strong>Australian</strong>, state and territory government health authorities and communicable<br />

disease control).<br />

• Zoster Immunoglobulin-VF (human) – CSL Limited. 160 mg/<br />

mL immunoglobulin (mainly IgG) prepared from human plasma<br />

containing high levels of antibody to the varicella-zoster virus. Single<br />

vials contain 200 IU of varicella-zoster antibody, with the actual<br />

volume stated on the label on the vial. Also contains glycine.<br />

High-titre zoster immunoglobulin (ZIG) is available from the <strong>Australian</strong> Red<br />

Cross Blood Service on a restricted basis for the prevention of varicella in highrisk<br />

subjects who report a significant exposure to varicella or HZ. ZIG has no<br />

proven use in the treatment of established varicella or zoster infection. ZIG is<br />

highly efficacious, but is often in short supply. Normal human immunoglobulin<br />

(NHIG) can be used for the prevention of varicella if ZIG is unavailable.<br />

Post-exposure prophylaxis using varicella vaccine may also be indicated, if<br />

vaccination is not contraindicated (see below).<br />

Zoster immunoglobulin should only be given by IM injection.<br />

‘Significant exposure’ to VZV is defined as living in the same household as a<br />

person with active varicella or HZ, or direct face-to-face contact with a person<br />

with varicella or HZ for at least 5 minutes, or being in the same room for at least<br />

1 hour. In the case of varicella infection, the period of infectivity is from 48 hours<br />

before the onset of rash until crusting of all lesions has occurred. Transmission<br />

from a person with localised zoster is less likely than from a person with<br />

varicella. 4<br />

Immunocompetent varicella contacts should be tested for varicella-zoster<br />

antibodies.<br />

ZIG must be given early in the incubation period (within 96 hours of exposure),<br />

but may have some efficacy if administered out to as late as 10 days post<br />

exposure. ZIG is able to prevent or ameliorate varicella in infants


should be tested for varicella-zoster antibodies following contact with a person<br />

with confirmed varicella. However, testing should not delay ZIG administration<br />

after initial exposure to a case. 84-86<br />

ZIG administration (preferably within 96 hours and up to 10 days after exposure)<br />

is required for the following groups and should not be delayed by testing (if<br />

indicated below):<br />

• Pregnant women who are presumed to be susceptible to varicella infection.<br />

If practicable, they should be tested for varicella-zoster antibodies before<br />

ZIG is given. 5<br />

• Neonates whose mothers develop varicella from 7 or fewer days before<br />

delivery to 2 days after delivery. ZIG must be given, as the neonatal mortality<br />

without ZIG is up to 30% in this setting. 1,7 ZIG must be given as early as<br />

possible in the incubation period.<br />

• Neonates exposed to varicella in the 1st month of life, if the mother has no<br />

personal history of infection with VZV and is seronegative. 27 ZIG should<br />

be given, due to the increased risk of severe varicella in newborns of<br />

seronegative women.<br />

• Premature infants (born at


high-dose intravenous NHIG are likely to be protected and probably do not<br />

require ZIG if the last dose of NHIG was given 3 weeks or less before exposure.<br />

Vaccination for post-exposure prophylaxis<br />

If VV is not contraindicated, it can be offered to non-immune age-eligible<br />

children and adults who have a significant exposure to varicella or HZ and<br />

wish to be protected against primary infection with VZV (see ‘Post-exposure<br />

vaccination’ in 4.22.7 Recommendations above). 51-55 Vaccination has the added<br />

benefit of reducing the likelihood of varicella infection, particularly moderate<br />

to severe disease, following exposure, and also provides long-term protection.<br />

Vaccination of exposed persons during outbreaks has also been shown to prevent<br />

further cases and to control outbreaks. 55 If MMR vaccination is also indicated,<br />

MMRV vaccine can be used in children


MMRV vaccine should not be used routinely as the 1st dose of MMR-containing<br />

vaccine in children aged


4.23 YELLOW FEVER<br />

4.23.1 Virology<br />

Yellow fever is a viral haemorrhagic fever caused by an RNA flavivirus that<br />

is transmitted by mosquitoes. Aedes aegypti, a highly domesticated mosquito<br />

found throughout the tropics, is the vector responsible for person-to-person<br />

transmission of the yellow fever virus in urban and inhabited rural areas in<br />

endemic countries.<br />

4.23.2 Clinical features<br />

<strong>The</strong> clinical spectrum of yellow fever varies from a non-specific febrile illness to<br />

fatal haemorrhagic fever. 1 After an incubation period of 3 to 6 days, the disease<br />

begins abruptly with fever, prostration, myalgia and headache. <strong>The</strong> patient<br />

appears acutely ill with congestion of the conjunctivae; there is an intense<br />

viraemia during this ‘period of infection’, which lasts 3 to 4 days. 1 This may be<br />

followed by the ‘period of remission’, in which the fever and symptoms settle<br />

over 24 to 48 hours, during which the virus is cleared by immune responses. 1<br />

Approximately 15 to 25% of patients may then relapse with a high fever,<br />

vomiting, epigastric pain, jaundice, renal failure and haemorrhage: ‘the period<br />

of intoxication’. 1 <strong>The</strong>se complications can be severe, and reflect the viscerotropic<br />

nature of the yellow fever virus (its ability to infect the liver, heart and<br />

kidneys). <strong>The</strong> case-fatality rate varies widely, but can be more than 20% in local<br />

populations. 2<br />

4.23.3 Epidemiology<br />

Yellow fever occurs in tropical regions of Africa and Central and South America.<br />

In both regions the virus is enzootic in rainforest monkeys and canopy mosquito<br />

species; sporadic human cases occur when people venture into these forests<br />

(‘sylvatic or jungle yellow fever’). 1<br />

In moist savannah regions in Africa, especially those adjacent to rainforests, tree<br />

hole-breeding Aedes mosquito species are able to transfer yellow fever virus from<br />

monkeys to people and then between people, leading to small-scale outbreaks<br />

(‘intermediate yellow fever’).<br />

Ae. aegypti occurs in both heavily urbanised areas and settled rural areas in<br />

tropical Africa and the Americas. 1 Epidemics of ‘urban yellow fever’ occur when<br />

a viraemic individual (with yellow fever) infects local populations of Ae. aegypti;<br />

such epidemics can be large and very difficult to control. Although Ae. aegypti<br />

also occurs throughout much of tropical Asia and Oceania (including north<br />

Queensland), yellow fever has never been reported from these regions.<br />

Although yellow fever is undoubtedly markedly under-reported, it is clear that<br />

there has been a considerable increase in the reported number of outbreaks, and<br />

therefore cases, of yellow fever in past decades. 3 Most of this increase was in<br />

PART 4 VACCINE-PREVENTABLE DISEASES 439<br />

4.23 YELLOW FEVER


Africa, particularly in West African countries. 3,4 In 2008, there were 24 cases of<br />

yellow fever reported near Asunción in Paraguay with 8 deaths. 5<br />

<strong>The</strong> risk of susceptible travellers acquiring yellow fever varies considerably<br />

with season, location, duration of travel and utilisation of mosquito avoidance<br />

measures. <strong>The</strong>re have been reported cases of yellow fever, all fatal, in<br />

unvaccinated travellers to Africa and South America. 6<br />

4.23.4 Vaccine<br />

• Stamaril – Sanofi Pasteur Pty Ltd (live attenuated yellow fever virus<br />

[17D strain]). Lyophilised powder in a monodose vial with a pre-filled<br />

diluent syringe. Each 0.5 mL reconstituted dose contains ≥1000 mouse<br />

LD 50 units; 16.0 mg lactose; 8.0 mg sorbitol; 0.833 mg L-histidine<br />

hydrochloride. May contain traces of egg proteins.<br />

Yellow fever vaccine is a live, freeze-dried preparation of attenuated 17D strain<br />

yellow fever virus cultured in, and harvested from, embryonated chicken eggs.<br />

<strong>The</strong> vaccine does not contain antibiotics, preservatives or gelatin.<br />

Vaccination elicits protective levels of neutralising antibodies in approximately<br />

90% of adult vaccine recipients by day 14, and in virtually all by day 28. 7<br />

Immunity is long-lasting and perhaps life-long; regardless, revaccination is<br />

required after 10 years under International Health Regulations for a valid<br />

International Certificate of Vaccination or Prophylaxis (ICVP) against yellow<br />

fever. Because the vaccine produces a transient very low level viraemia in healthy<br />

adult recipients, they cannot serve as a source of infection for mosquitoes. 7<br />

Although the efficacy of the yellow fever vaccine has never been determined in<br />

prospective clinical trials, there is considerable observational evidence that it is<br />

very effective in preventing the disease. 7<br />

4.23.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 8 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

Stamaril must be reconstituted by adding the entire contents of the diluent syringe<br />

to the vial and shaking until the powder is completely dissolved. Reconstituted<br />

vaccine must be used within 1 hour.<br />

4.23.6 Dosage and administration<br />

<strong>The</strong> dose of yellow fever vaccine for children and adults is 0.5 mL, to be given by<br />

either IM or SC injection.<br />

Test doses of yellow fever vaccine should never be used (see 4.23.13 Variations<br />

from product information below).<br />

440 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Co-administration with other vaccines<br />

If administration of both yellow fever and other parenteral live viral vaccines is<br />

indicated, the vaccines should be given either on the same day or at least 4 weeks<br />

apart (see 4.23.10 Precautions below).<br />

Inactivated vaccines and oral live vaccines relevant to travel (e.g. cholera,<br />

typhoid) can be given with, or at any time before or after, yellow fever vaccine.<br />

Yellow fever vaccine can be given at the same time as the Imojev Japanese<br />

encephalitis vaccine, 9 using separate syringes and separate injection sites.<br />

4.23.7 Recommendations<br />

Children aged


are free to set their own requirements for entry and some countries require a<br />

valid International Certificate of Vaccination or Prophylaxis against yellow<br />

fever or a valid letter of exemption for all arriving travellers. A country may<br />

require such documentation even for travellers who are only in transit through<br />

that country. <strong>The</strong> most recent WHO list of individual country yellow fever<br />

vaccination requirements and recommendations for travellers can be found at<br />

www.who.int/ith/chapters/ith2012en_countrylist.pdf. As yellow fever disease<br />

patterns, like other diseases, are constantly changing, it is recommended that<br />

the entry requirements for yellow fever vaccination for the countries a traveller<br />

intends to enter or transit through be confirmed by contacting the country’s<br />

foreign missions in Australia.<br />

Travellers >1 year of age entering or returning to Australia within 6 days of<br />

leaving a country on Australia’s list of yellow fever declared places are required<br />

to have a valid International Certificate of Vaccination or Prophylaxis with<br />

proof of valid yellow fever vaccination (see below). This list is developed based<br />

on the WHO list of countries with risk of yellow fever virus transmission and<br />

international surveillance data, and is available from the <strong>Australian</strong> Government<br />

Department of Health and Ageing’s yellow fever fact sheet (www.health.gov.au/<br />

yellowfever). Travellers who do not have a valid certificate are provided with<br />

information on yellow fever and required to promptly seek medical assessment if<br />

they develop relevant symptoms within 6 days of leaving the declared place.<br />

Yellow fever vaccine can be administered only by Yellow Fever Vaccination<br />

Centres approved by the relevant state or territory health authorities. Each<br />

yellow fever vaccination is to be recorded in an International Certificate of<br />

Vaccination or Prophylaxis, with proof of valid yellow fever vaccine; the<br />

certificate must include the vaccinated person’s name and signature (or the<br />

signature of a parent or guardian of a child), and the signature of a person<br />

approved by the relevant health authority. <strong>The</strong> date of the vaccination must be<br />

recorded in day–month–year sequence, with the month written in letters, and the<br />

official stamp provided by the state or territory health authority must be used.<br />

<strong>The</strong> certificate becomes valid 10 days after vaccination, and remains valid for<br />

10 years.<br />

Note: People with a true contraindication to yellow fever vaccine (see 4.23.9<br />

Contraindications below) who intend to travel to yellow fever risk countries<br />

should obtain a letter from a doctor, clearly stating the reason for withholding<br />

the vaccine. <strong>The</strong> letter should be formal, signed and dated, and on the practice’s<br />

letterhead. Arriving travellers who possess an exemption from the yellow fever<br />

vaccination are provided with information on yellow fever and required to<br />

promptly seek medical assessment if they develop relevant symptoms.<br />

442 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


4.23.8 Pregnancy and breastfeeding<br />

Yellow fever vaccine is not recommended for pregnant women or women<br />

breastfeeding infants aged


Altered immune status<br />

As with all live viral vaccines, the yellow fever vaccine should generally not<br />

be given to people who are immunocompromised due to either disease or<br />

medical treatment (see 3.3.3 Vaccination of immunocompromised persons). However,<br />

studies of small numbers of HIV-infected participants, mainly travellers with<br />

CD4 + counts >200 per μL, have shown reduced immune response, but good<br />

tolerability. 15<br />

Thymus disorders<br />

People with a history of any thymus disorder, including myasthenia gravis,<br />

thymoma, thymectomy and DiGeorge syndrome, or thymic damage from<br />

chemoradiotherapy or graft-versus-host disease, should not be given the yellow<br />

fever vaccine due to the increased risk of yellow fever vaccine-associated<br />

viscerotropic disease (see 4.23.11 Adverse events below).<br />

4.23.10 Precautions<br />

Adults aged ≥60 years<br />

<strong>The</strong> risk of severe adverse events following yellow fever vaccine is considerably<br />

greater in those aged ≥60 years than in younger adults. 16-19<br />

Adults ≥60 years of age should be given yellow fever vaccine only if they intend<br />

to travel to endemic countries (as recommended above) and they have been<br />

informed about the (albeit very low) risks of developing a severe complication.<br />

Vaccination with other live attenuated parenteral vaccines<br />

<strong>The</strong> administration of other parenteral live viral vaccines (e.g. MMR, MMRV,<br />

varicella or zoster vaccine) should be on the same day as yellow fever vaccine, or<br />

separated by a 4-week interval.<br />

4.23.11 Adverse events<br />

Mild adverse events<br />

Adverse events following yellow fever vaccine are generally mild. Vaccine<br />

recipients often report mild headaches, myalgia and low-grade fevers or other<br />

minor symptoms for 5 to 10 days post vaccination. In clinical trials in which<br />

symptoms are actively elicited, up to 25% of vaccine recipients report mild<br />

adverse events and up to 1% curtail regular activities. 7,19,20<br />

Immediate hypersensitivity reactions<br />

Immediate hypersensitivity reactions, including anaphylaxis, following yellow<br />

fever vaccine are very rare, with an incidence of less than 1 in 1 million, and<br />

occur principally in people with anaphylactic sensitivity to eggs. 7,18,19 Although<br />

it has been suggested that an anaphylactic sensitivity to gelatin (added as a<br />

stabiliser to some yellow fever vaccines) may also precipitate anaphylaxis<br />

following vaccination, 21 Stamaril does not contain gelatin.<br />

444 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Vaccine-associated neurotropic adverse events<br />

Yellow fever vaccine-associated neurotropic disease (YF-AND) is rare. 18<br />

At least 25 cases of meningoencephalitis following yellow fever<br />

vaccination have been reported. 7 However, 15 of these cases occurred in<br />

the 1950s in infants ≤7 months of age. Following recommendations in<br />

the early 1960s not to vaccinate young infants, the incidence of vaccineassociated<br />

meningoencephalitis declined considerably. 7 Nevertheless,<br />

these adverse events, albeit very rare, still occur in adults; the risk is<br />

greater in persons ≥60 years of age. 16,17<br />

Vaccine-associated viscerotropic adverse events<br />

Recently, an apparently very rare (and often fatal) complication, yellow fever<br />

vaccine-associated viscerotropic disease (YF-AVD), characterised by multiorgan<br />

system failure, has been recognised following yellow fever vaccination. It<br />

appears that overwhelming infection with the 17D vaccine virus is responsible<br />

for these viscerotropic adverse events. 7,18<br />

Vaccine-associated viscerotropic adverse events do not appear to be caused by<br />

altered virulence of the vaccine virus, but rather appear to be related to host<br />

factors. Although cases have occurred in younger persons, it is apparent that the<br />

risk is increased with advanced age, particularly in those aged ≥60 years. 16,17,19<br />

Another host factor associated with an increased risk of a viscerotropic adverse<br />

event is pre-existing thymus disease. Published reports of YF-AVD cases have<br />

indicated that 4 of the 27 reported cases had a history of thymic tumour and<br />

thymectomy, both uncommon conditions. 18,22<br />

4.23.12 Public health management of yellow fever<br />

Yellow fever is a notifiable and quarantinable disease in all states and territories<br />

in Australia.<br />

Further instructions about the public health management of yellow fever,<br />

including management of cases of yellow fever and their contacts, should<br />

be obtained from state/territory public health authorities (see Appendix 1<br />

Contact details for <strong>Australian</strong>, state and territory government health authorities and<br />

communicable disease control).<br />

4.23.13 Variations from product information<br />

<strong>The</strong> product information states that pregnancy is a contraindication to the yellow<br />

fever vaccine. <strong>The</strong> ATAGI recommends instead that pregnant women who insist<br />

on travelling to endemic countries should be vaccinated.<br />

<strong>The</strong> product information suggests that a 0.1 mL test dose of yellow fever vaccine<br />

can be used intradermally to assess an individual with suspected allergy to the<br />

vaccine. <strong>The</strong> ATAGI instead recommends that (with the exception of Q fever<br />

vaccine) test doses should never be used.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

PART 4 VACCINE-PREVENTABLE DISEASES 445<br />

4.23 YELLOW FEVER


4.24 ZOSTER (herpes zoster)<br />

4.24.1 Virology<br />

Varicella-zoster virus (VZV) is a DNA virus that is a member of the herpesvirus<br />

family. Primary infection with VZV is known as varicella or ‘chickenpox’. 1<br />

Herpes zoster (HZ), or ‘shingles’, is caused by reactivation of latent VZV, which<br />

typically resides in the dorsal root or trigeminal nerve ganglia following primary<br />

infection. 1<br />

4.24.2 Clinical features<br />

Reactivation of VZV causing HZ is thought to be particularly due to a decline in<br />

cellular immunity to the virus, and presents clinically as a unilateral vesicular<br />

rash in a dermatomal distribution in the majority of cases. A prodromal phase<br />

occurs 48 to 72 hours prior to the appearance of the lesions in 80% of cases. 2<br />

Associated symptoms may include headache, photophobia, malaise, and an<br />

itching, tingling or severe pain in the affected dermatome. 3,4 In the majority of<br />

patients, HZ is an acute and self-limiting disease, with the rash lasting 10 to<br />

15 days. 1,3 However, complications can occur, especially with increasing age.<br />

Post-herpetic neuralgia (PHN), the most frequent debilitating complication<br />

of HZ, is a neuropathic pain syndrome that persists or develops after<br />

the dermatomal rash has healed. By definition, PHN is established when<br />

pain persists for longer than 3 months after the onset of the rash. 5,6 Other<br />

complications may occur, depending on the site of reactivation. <strong>The</strong>se include<br />

ophthalmic disease (such as keratitis and chorioretinitis), neurological<br />

complications (such as meningoencephalitis and myelitis), secondary bacterial<br />

skin infection, scarring and pneumonia. 7 Rarely, disseminated HZ may develop,<br />

with widespread vesicular rash, and visceral, central nervous system and<br />

pulmonary involvement. Disseminated disease is more common in persons who<br />

are immunocompromised. 4 Dermatomal pain without the appearance of rash is<br />

also documented (zoster siné herpéte).<br />

Antiviral therapy, if initiated within 3 days of the onset of HZ, has been shown<br />

to reduce the severity and duration of HZ and may reduce the risk of developing<br />

PHN. 8-12 However, despite medical therapy, PHN may persist for years and can<br />

be refractory to treatment. 13<br />

4.24.3 Epidemiology<br />

HZ occurs most commonly with increasing age (>50 years), immunocompromise,<br />

and following a history of varicella in the 1st year of life. <strong>The</strong> lifetime risk of<br />

reactivation of VZV causing HZ is estimated to be approximately 20 to 30%<br />

and it affects half of those who live to 85 years. 1,14-16 Second attacks of HZ occur<br />

in approximately 5% of immunocompetent persons, but are more frequent<br />

in persons who are immunocompromised. 3,17,18 Using <strong>Australian</strong> general<br />

practice and other data, approximately 490 cases per 100 000 (range 330–830<br />

per 100 000) are estimated to occur annually in all ages, with approximately<br />

446 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


1000 cases per 100 000 population in persons aged ≥50 years. 19-22 In the large<br />

efficacy study of zoster vaccine in the United States, the incidence of HZ in<br />

unimmunised participants ≥60 years of age was 1112 cases per 100 000 personyears.<br />

23 <strong>The</strong> incidence in persons aged 50–59 years is lower, with one study<br />

estimating a rate of 470 per 100 000 person-years. 24 <strong>The</strong> risk of HZ increases<br />

with immunocompromise; for example, rates of HZ are up to 15 times higher in<br />

those who are immunocompromised due to HIV infection, and, in the 1st year<br />

following haematopoietic stem cell transplantation (HSCT), up to 30% of patients<br />

may develop HZ. 3,25<br />

Overall, an estimated 13 to 26% of patients with HZ develop complications.<br />

Complications occur more frequently with increasing age, and with<br />

immunocompromise. 26,27 PHN occurs infrequently in young people, but, in<br />

patients over the age of 50 years, it complicates HZ in 25 to 50% of cases. 26,27<br />

Modelling studies of the impact of universal childhood vaccination programs<br />

for varicella have predicted that a rise in the incidence of HZ could occur, based<br />

on the assumption that exposure to wild-type VZV circulating in the community<br />

boosts immunity. 28 However, to date, multiple studies and surveillance data do<br />

not demonstrate any consistent changes in overall HZ incidence in the United<br />

States, which has a universal varicella vaccination program that commenced<br />

in 1995. 29-31 <strong>Australian</strong> data show an increase in HZ GP consultation rates<br />

over time, commencing prior to varicella vaccine introduction, which is<br />

likely to be due to the increasing age of the population. Age-standardised HZ<br />

hospitalisation rates have not declined since introduction of varicella vaccine,<br />

and the use of zoster vaccine has not yet been extensive enough in any country<br />

to expect an impact on HZ epidemiology. 32-34 In the United States, the incidence<br />

of HZ in children 50 years of age. It is important to note<br />

that the registered varicella vaccines are not indicated for use in preventing<br />

HZ in older people and Zostavax is not indicated for use in younger people<br />

who have not been previously immunised or infected with VZV. Zostavax is<br />

not indicated for use for therapeutic benefit during an acute HZ episode, nor<br />

for the treatment of PHN.<br />

A single large, randomised, double-blind, placebo-controlled efficacy study of<br />

the frozen formulation of Zostavax (known as the ‘Shingles Prevention Study’<br />

PART 4 VACCINE-PREVENTABLE DISEASES 447<br />

4.24 ZOSTER (HERPES ZOSTER)


[SPS]) was conducted among 38 546 adults aged ≥60 years and demonstrated<br />

that Zostavax significantly reduced the likelihood of developing both HZ and<br />

PHN. 23 Vaccination reduced the incidence of HZ by 51.3%, the incidence of PHN<br />

by 66.5%, and the burden of illness associated with HZ by 61.1% over a median<br />

of more than 3 years follow-up. 23 <strong>The</strong> vaccine was more efficacious in reducing<br />

HZ in persons aged 60–69 years than in those aged 70–79 years (64% compared<br />

with 41% efficacy). However, efficacy against PHN was similar in both age<br />

groups. 23 Efficacy against HZ in the ≥80 years age group was lower (18% and not<br />

statistically different to placebo). However, there were fewer participants of this<br />

age in the SPS. 36 In those who developed HZ despite vaccination, the severity<br />

of pain associated with the episode was also reduced. 37 Another randomised<br />

controlled study in >22 000 50–59-year olds demonstrated a reduction in the<br />

incidence of HZ after a follow-up period of 1 year or more, with a vaccine<br />

efficacy for preventing HZ of 69.8%. 38 In these clinical trials many participants<br />

were treated with antiviral and pain medication for their HZ, suggesting that<br />

the effect of the vaccine was in addition to any benefit obtained from medical<br />

therapy. 23,38 Efficacy of a single dose of zoster vaccine appears to decline over<br />

time, with recent data suggesting persistent efficacy through to the 5th year post<br />

vaccination, with uncertain efficacy beyond that point. 39 <strong>The</strong> need for a booster<br />

dose has not yet been determined.<br />

<strong>The</strong> Shingles Prevention Study, together with other smaller studies,<br />

demonstrated that Zostavax is safe and generally well tolerated among adults<br />

≥50 years of age. 23,38 In the SPS, the most common adverse events were injection<br />

site reactions, with Zostavax more likely to result in erythema, pain and swelling<br />

at the injection site than placebo (48% versus 17%, respectively). Varicellalike<br />

rashes at the injection site were also more common in vaccine recipients;<br />

however, varicella-like rash not localised to the injection site did not occur more<br />

often. Varicella- or zoster-like rashes that were PCR-positive for VZV were<br />

mostly due to wild-type VZV. 23 Fever was no more common in vaccine recipients;<br />

however, the rate of vaccine-related systemic symptoms was higher (Zostavax<br />

6.3% versus placebo 4.9%), with the most frequently reported systemic symptoms<br />

being headache and fatigue. 23 Mild to moderate adverse events, particularly<br />

injection site reactions, were higher in vaccine recipients aged 50–59 years than in<br />

those aged ≥60 years. 38,40<br />

In Australia, a refrigerated form of Zostavax is registered on the basis of<br />

comparable immunogenicity and safety to the frozen vaccine formulation that<br />

was used in the SPS. 41 Zostavax was registered for use in persons 50–59 years<br />

of age based on a study that demonstrated similar immunogenicity in this age<br />

group compared with those ≥60 years of age, 40 and has since been shown to<br />

reduce the incidence of HZ in this population. 38 A study of the simultaneous<br />

administration of Zostavax with inactivated influenza vaccine (given separately<br />

and at different injection sites) demonstrated comparable immunogenicity and<br />

safety to giving the vaccines at different times. 42 A study of the simultaneous<br />

448 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


administration of Zostavax with 23-valent pneumococcal polysaccharide vaccine<br />

(Pneumovax 23; 23vPPV) suggested that the immunogenicity of Zostavax was<br />

reduced when administered simultaneously with Pneumovax 23, compared with<br />

administration 4 weeks apart. 43 <strong>The</strong> immunogenicity of Pneumovax 23 was not<br />

affected. However, an observational study from the United States suggests this<br />

may not impact on Zostavax effectiveness. 44-46<br />

Zostavax availability in Australia has been limited due to manufacturer supply<br />

constraints.<br />

• Zostavax – CSL Limited/Merck & Co Inc (live attenuated Oka/Merck<br />

strain of varicella-zoster virus). Lyophilised powder in a monodose<br />

vial with separate diluent. Each 0.65 mL reconstituted dose contains<br />

≥19 400 plaque-forming units of attenuated varicella-zoster virus;<br />

sucrose; gelatin; urea; monosodium glutamate; residual components<br />

of MRC-5 cells; traces of neomycin and bovine serum albumin.<br />

4.24.5 Transport, storage and handling<br />

Transport according to National vaccine storage guidelines: Strive for 5. 47 Store at<br />

+2°C to +8°C. Do not freeze. Protect from light.<br />

Zostavax is less stable than other commonly used live viral vaccines, and<br />

adherence to storage and reconstitution requirements is very important.<br />

Zostavax must be reconstituted by adding the entire contents of the diluent<br />

container to the vial and shaking until the powder is completely dissolved.<br />

Reconstitute immediately upon removal from the refrigerator. Reconstituted<br />

vaccine must be used within 30 minutes.<br />

4.24.6 Dosage and administration<br />

<strong>The</strong> dose of Zostavax is 0.65 mL, to be given by SC injection.<br />

Zostavax must never be given where varicella (chickenpox) vaccine is indicated.<br />

Zoster vaccine is only registered for use in adults ≥50 years of age.<br />

Co-administration with other vaccines<br />

Zostavax can be given at the same time as influenza vaccine, 42 using separate<br />

syringes and injection sites.<br />

Simultaneous administration of Zostavax with pneumococcal polysaccharide<br />

vaccine is not routinely recommended; if possible the two vaccines should be<br />

given at least 4 weeks apart. However, inadvertent administration of Zostavax<br />

and pneumococcal polysaccharide vaccine at the same time or at an interval of<br />

less than 4 weeks does not require revaccination (see 4.24.4 Vaccine above).<br />

PART 4 VACCINE-PREVENTABLE DISEASES 449<br />

4.24 ZOSTER (HERPES ZOSTER)


Zostavax can be administered at the same visit as, or at any time following<br />

receipt of, other inactivated vaccines (e.g. tetanus-containing vaccines), if<br />

required.<br />

If administration of both Zostavax and another live parenteral vaccine (e.g. MMR<br />

or yellow fever) is indicated, the vaccines should be given either on the same day<br />

or at least 4 weeks apart. (See also 4.22 Varicella.)<br />

4.24.7 Recommendations<br />

Adults aged ≥60 years<br />

A single dose of zoster vaccine is recommended for adults ≥60 years of age who<br />

have not previously received a dose of zoster vaccine. Routine serological testing<br />

prior to receipt of zoster vaccine is not indicated and it is not necessary to elicit<br />

a history of previous varicella (chickenpox) infection (see ‘Serological testing<br />

before and after zoster vaccination’ below).<br />

Persons with chronic medical conditions, such as arthritis, chronic renal<br />

failure, diabetes and other conditions, can be given zoster vaccine, unless a<br />

contraindication or precaution exists due to their condition or medical treatment<br />

(see 4.24.9 Contraindications and 4.24.10 Precautions below). Persons with<br />

significant immunocompromise should not receive zoster vaccine (see also 3.3.3<br />

Vaccination of immunocompromised persons).<br />

<strong>The</strong> zoster vaccine has been shown to be less efficacious in persons aged ≥80<br />

years and may be less likely to provide a clinical benefit in this age group (see<br />

4.24.4 Vaccine above).<br />

Adults aged 50–59 years<br />

Routine population-based use of zoster vaccine in persons aged 50–59 years is<br />

not recommended. Although the incidence of HZ in persons 50–59 years of age<br />

is higher than in younger age groups, 19,22 and zoster vaccine is efficacious in<br />

50–59-year olds, 38 the likelihood of developing PHN and other complications<br />

of HZ is lower in this age group than in those ≥60 years of age. 24,48 Persons aged<br />

50–59 years who wish to protect themselves against HZ can be vaccinated;<br />

however, the duration of efficacy, and need for a booster dose at a later age, is<br />

not yet determined (see 4.24.4 Vaccine above).<br />

Persons aged


diagnosis. In addition, the risk of a repeat episode of zoster has been estimated<br />

at approximately 5% in immunocompetent persons. 17,18,48 Persons with a history<br />

of HZ were excluded from the SPS, so no data on the efficacy of the vaccine in<br />

those with a history of HZ is available. <strong>The</strong> safety and immunogenicity of zoster<br />

vaccine in persons with a history of HZ has been studied in one small clinical<br />

trial; the vaccine was well tolerated and immunogenic. 49 Injection site reactions<br />

were more common in vaccine recipients than in placebo recipients, but similar<br />

to vaccine recipients in the SPS. Systemic adverse events were similar between<br />

groups. 23 <strong>The</strong> length of time following an episode of HZ after which it would be<br />

reasonable to vaccinate has not been established. However, it is suggested that<br />

the vaccine could be given at least 1 year after the episode of HZ.<br />

Persons previously vaccinated with varicella vaccine<br />

Zoster vaccination of persons who have previously received varicella vaccine is<br />

not recommended at this time. <strong>The</strong>re have been limited studies of the safety and<br />

immunogenicity of zoster vaccine in this setting, and the currently available data<br />

are insufficient to suggest a benefit from vaccination. It is not yet known whether,<br />

in the future, populations vaccinated with varicella vaccine will experience rates<br />

of HZ sufficient to warrant zoster vaccination. Preliminary information suggests<br />

that the incidence of HZ in persons who have received varicella vaccine is lower<br />

than in those infected with wild-type varicella. 30<br />

Persons with immunocompromise due to HIV/AIDS<br />

Studies of the use of zoster vaccine in HIV-infected persons have not been<br />

completed. However, persons with asymptomatic HIV infection may be<br />

considered for vaccination on a case-by-case basis after seeking appropriate<br />

specialist advice. Serological confirmation of previous VZV infection must be<br />

obtained prior to vaccination (see ‘Serological testing before and after zoster<br />

vaccination’ below). Although asymptomatic HIV-infected persons are likely<br />

to have a higher relative risk of developing HZ in the future, 25 it is possible that<br />

both the efficacy and the safety of zoster vaccination may be reduced in such<br />

recipients, as compared with uninfected persons.<br />

Vaccination with zoster vaccine is not recommended for persons with AIDS or<br />

symptomatic HIV infection (see 3.3 Groups with special vaccination requirements,<br />

Table 3.3.4 Categories of immunocompromise in HIV-infected persons, based on<br />

age-specific CD4 + counts and percentage of total lymphocytes), or significant<br />

immunocompromise due to other diseases and/or treatment (see 4.24.9<br />

Contraindications below).<br />

Persons anticipating future significant immunocompromise<br />

Immunocompetent persons who anticipate alteration of their immunity because<br />

of their existing illness can be given zoster vaccine on a case-by-case basis<br />

after seeking appropriate specialist advice. 36 Persons with conditions such as<br />

chronic lymphocytic leukaemia, conditions requiring organ transplantation, 50<br />

PART 4 VACCINE-PREVENTABLE DISEASES 451<br />

4.24 ZOSTER (HERPES ZOSTER)


solid tumours that will require future chemotherapy or radiation therapy, and<br />

inflammatory diseases (e.g. rheumatoid arthritis, systemic lupus erythematosus,<br />

inflammatory bowel disease, psoriasis) may have minimal alteration to their<br />

immune system, but can anticipate significant immunocompromise in the future<br />

due to their disease and/or treatment. Since these persons are at higher risk of<br />

developing zoster than if they remained immunocompetent, vaccination at least<br />

1 month prior to the onset of immunocompromise may be appropriate (after<br />

seeking specialist advice). 36 Serological confirmation of previous VZV infection<br />

must be obtained prior to vaccination (see ‘Serological testing before and after<br />

zoster vaccination’ below).<br />

Household contacts of persons who are immunocompromised<br />

Vaccination is recommended for persons ≥50 years of age who are household<br />

contacts of a person who is immunocompromised. Based on evidence that the<br />

rate of VZV-like rashes after vaccination is extremely low, it is unlikely that<br />

transmission of vaccine-associated virus to a susceptible contact would occur. 23<br />

If a vaccinated person develops a varicella- or zoster-like rash, they should cover<br />

the rash and avoid contact with persons who are immunocompromised for the<br />

duration of the rash. <strong>The</strong> efficacy of the HZ vaccine is less than 100%, and rashes<br />

in vaccinated persons may be due to reactivation of wild-type VZV. If household<br />

contacts (


Laboratory testing to check for an immune response after zoster vaccination<br />

is not recommended. Zoster vaccine boosts both humoral (IgG) and cellular<br />

immune responses; however, confirmation of such immune responses is neither<br />

necessary nor predictive of protection against the development of zoster.<br />

4.24.8 Pregnancy and breastfeeding<br />

VZV-containing vaccines are contraindicated in pregnant women, although<br />

women of child-bearing age are not eligible for zoster vaccination. Pregnancy<br />

should be avoided for 28 days after vaccination (see 4.22 Varicella).<br />

A non-immune pregnant household contact is not a contraindication to zoster<br />

vaccination.<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

4.24.9 Contraindications<br />

Anaphylaxis to vaccine components<br />

Zoster vaccine is contraindicated in persons who have had:<br />

• anaphylaxis following a previous dose of any VZV-containing vaccine<br />

• anaphylaxis following any vaccine component.<br />

Persons who are immunocompromised<br />

Live attenuated zoster vaccine is contraindicated in persons with significant<br />

immunocompromise due to either a primary or acquired medical condition, or<br />

due to medical treatment. This includes persons receiving high-dose systemic<br />

immunosuppressive therapy, such as chemotherapy, radiation therapy or<br />

oral corticosteroids; persons suffering from malignant conditions of the<br />

reticuloendothelial system (such as lymphoma, leukaemia, Hodgkin’s disease);<br />

and any person with similar immunocompromise due to a disease or treatment<br />

(see 3.3.3 Vaccination of immunocompromised persons).<br />

Persons who have been receiving high-dose systemic steroids (or equivalent)<br />

and have ceased therapy may be vaccinated (see 3.3.3 Vaccination of<br />

immunocompromised persons). Persons on low-dose corticosteroids or less<br />

significantly immunocompromised than described above, may be considered for<br />

vaccination on a case-by-case basis. Studies of zoster vaccine in such persons are<br />

being conducted.<br />

4.24.10 Precautions<br />

Vaccination with other live attenuated parenteral vaccines<br />

If zoster vaccine is to be given around the same time as another live viral<br />

parenteral vaccine (e.g. MMR, yellow fever), the vaccines should be given either<br />

at the same visit or at least 4 weeks apart.<br />

PART 4 VACCINE-PREVENTABLE DISEASES 453<br />

4.24 ZOSTER (HERPES ZOSTER)


Vaccination before or after immunoglobulin or blood product administration<br />

Zoster vaccine can be given at any time before or after administration of<br />

immunoglobulin or any antibody-containing blood product. This is because<br />

zoster vaccine is indicated in persons who, because of their age, are assumed to<br />

have had a previous VZV infection and, therefore, already have serum antibody<br />

levels comparable to those found in blood products. (See also 3.3.4 Vaccination of<br />

recent recipients of normal human immunoglobulin and other blood products.)<br />

Persons receiving long-term aspirin or salicylate therapy<br />

Persons receiving long-term salicylate therapy (aspirin) can be vaccinated if<br />

indicated. <strong>The</strong>re have been no reports of an association between Reye syndrome<br />

and varicella vaccination, and it is unlikely that vaccination of a previously VZVinfected<br />

older person with zoster vaccine carries any risk of Reye syndrome.<br />

Persons receiving antiviral medication<br />

It is possible that the use of antivirals with anti-VZV activity, such as acyclovir,<br />

famciclovir or valaciclovir, may interfere with the replication of the Zostavax<br />

live attenuated virus. Persons on such antiviral medication should cease<br />

treatment no less than 24 hours prior to vaccination and for at least 14 days after<br />

vaccination. 36,53<br />

4.24.11 Adverse events<br />

Injection site reactions (including erythema, pain, swelling and/or itch at the<br />

injection site) occurred in approximately half of clinical trial participants given<br />

Zostavax, irrespective of a previous history of HZ (see also 4.24.4 Vaccine above).<br />

Varicella-like rashes at the injection site occurred rarely, in 0.1% of recipients;<br />

however, they were more common than in placebo recipients. Varicella-like<br />

rashes that were not localised to the injection site were also rare, and did not<br />

occur more often in vaccine compared with placebo recipients (0.1% in both<br />

groups). In the clinical trials in which rashes were analysed by PCR for VZV, the<br />

majority were due to wild-type virus; only 2 subjects were found to have rashes<br />

due to the Oka/Merck VZV vaccine strain (see also 4.24.4 Vaccine above).<br />

Fever >38.3°C was not seen more commonly in vaccine recipients, and occurred<br />

in


4.24.12 Variations from product information<br />

<strong>The</strong> product information for Zostavax states that the vaccine can be administered<br />

concurrently with inactivated influenza vaccine but not with 23vPPV. <strong>The</strong> ATAGI<br />

instead recommends that Zostavax may be administered concurrently with<br />

other vaccines as indicated. <strong>The</strong> ATAGI also recommends that if inadvertent<br />

concomitant administration of Zostavax and pneumococcal polysaccharide<br />

vaccine occurs, there is no need to revaccinate.<br />

<strong>The</strong> product information for Zostavax states that the safety and efficacy of<br />

Zostavax have not been established in adults with known HIV infection,<br />

with or without evidence of immunocompromise. <strong>The</strong> ATAGI recommends<br />

instead that Zostavax may be administered to HIV-infected persons without<br />

immunocompromise on a case-by-case basis, after seeking appropriate specialist<br />

advice, and following confirmation of pre-existing immunity to VZV.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

PART 4 VACCINE-PREVENTABLE DISEASES 455<br />

4.24 ZOSTER (HERPES ZOSTER)


PART 5 PASSIVE IMMUNISATION<br />

5.1 PASSIVE IMMUNISATION USING<br />

IMMUNOGLOBULIN PREPARATIONS<br />

Immunoglobulin preparations are used to provide passive immunisation, that<br />

is, the direct administration of antibodies to a non-immune person to provide<br />

immediate protection against infection or disease.<br />

Immunoglobulin infusions are also indicated for some immunocompromised<br />

persons who are antibody-deficient. In addition, immunoglobulins are also used<br />

in the treatment of a number of specific immune-mediated conditions in order<br />

to modulate the disease course. For further information regarding the use of<br />

intravenous immunoglobulins, refer to Criteria for the clinical use of intravenous<br />

immunoglobulin in Australia1 (www.nba.gov.au/ivig/index.html).<br />

It is important to recognise that separate immunoglobulin preparations<br />

are provided for intramuscular (IM) use and for intravenous (IV) use.<br />

<strong>The</strong>se have different properties, and the preparations should be given<br />

only by the recommended route. Administration of IM immunoglobulin<br />

by the IV route will lead to severe reactions. For more information<br />

on intravenous immunoglobulin, refer to Criteria for the clinical use of<br />

intravenous immunoglobulin in Australia. 1<br />

<strong>The</strong>re are two types of immunoglobulin:<br />

• normal human immunoglobulin<br />

• specific immunoglobulins.<br />

Normal human immunoglobulin (NHIG) is derived from the pooled plasma of<br />

blood donors. It contains antibody to microbial agents that are prevalent in the<br />

general population.<br />

Specific immunoglobulin preparations are obtained from pooled blood<br />

donations from patients convalescing from the relevant infection, donors recently<br />

vaccinated with the relevant vaccine, or those who, on screening, have been<br />

found to have sufficiently high antibody concentrations. <strong>The</strong>se blood-derived<br />

specific immunoglobulins therefore contain concentrations of antibody to an<br />

individual organism or toxin at a higher titre than would be present in normal<br />

immunoglobulin.<br />

Donors of blood used for the production of NHIG and specific immunoglobulin<br />

products are screened, and the products are treated to minimise the risk of<br />

456 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


the immunoglobulin preparations containing HIV, hepatitis A, hepatitis B or<br />

hepatitis C viruses, or parvovirus. Two dedicated pathogen inactivation steps<br />

are incorporated into the manufacturing process. A pasteurisation step is usually<br />

used during manufacture. <strong>The</strong> risk of prion transmission remains theoretical (see<br />

www.transfusion.com.au/adverse_events/risks/estimates for further details).<br />

5.1.1 Availability of immunoglobulins<br />

CSL Limited supplies NHIG for IM use both directly to hospitals and to<br />

the <strong>Australian</strong> Red Cross Blood Service. Rabies immunoglobulin, tetanus<br />

immunoglobulin and botulism antitoxin can only be obtained by application<br />

tostate/territory health authorities. Respiratory syncytial virus (RSV)<br />

monoclonal antibody (Synagis; Abbott Australia) is available commercially.<br />

Other specific immunoglobulins (for hepatitis B, cytomegalovirus, tetanus and<br />

varicella-zoster), which are derived from <strong>Australian</strong> donated plasma, can be<br />

obtained only from the <strong>Australian</strong> Red Cross Blood Service with permission from<br />

an <strong>Australian</strong> Red Cross Blood Service medical officer. <strong>The</strong> <strong>Australian</strong> Red Cross<br />

Blood Service supplies these products free of charge.<br />

<strong>The</strong> Blood Service can be contacted by telephone nationally on 13 14 95; callers<br />

will then be connected to the relevant state or territory <strong>Australian</strong> Red Cross<br />

Blood Service branch.<br />

Individual state or territory contact numbers:<br />

• <strong>Australian</strong> Capital Territory 02 6206 6024<br />

• New South Wales 1300 478 348<br />

• Northern Territory 08 8928 5116<br />

• Queensland 07 3838 9010<br />

• South Australia 08 8422 1201<br />

• Tasmania 03 6230 6209<br />

• Victoria 03 9694 0200<br />

• Western Australia 08 9421 2869<br />

5.1.2 Transport, storage and handling<br />

Store all immunoglobulins at +2°C to +8°C. Do not freeze. Protect from light.<br />

5.1.3 Normal human immunoglobulin for intramuscular use<br />

Normal human immunoglobulin (NHIG) is prepared by plasma fractionation<br />

of blood collected from volunteer donors by the <strong>Australian</strong> Red Cross Blood<br />

Service. It is a sterile solution of immunoglobulin, mainly IgG, and contains<br />

those antibodies commonly present in adult human blood. In Australia, NHIG is<br />

supplied as a 16% solution and made available through the <strong>Australian</strong> Red Cross<br />

Blood Service.<br />

PART 5 PASSIVE IMMUNISATION 457<br />

5.1 PASSIVE IMMUNISATION<br />

USING IMMUNOGLOBULIN<br />

PREPARATIONS


• Normal Immunoglobulin-VF (human) (NHIG; for intramuscular use)<br />

– CSL Limited. 160 mg/mL immunoglobulin (mainly IgG) prepared<br />

from <strong>Australian</strong> blood donations. Supplied in 2 mL and 5 mL vials.<br />

Also contains glycine.<br />

Administration<br />

NHIG should be given by deep IM injection, using an appropriately sized<br />

needle. <strong>The</strong> NHIG should be introduced slowly into the muscle, to reduce pain.<br />

This product must not be administered intravenously because of possible severe<br />

adverse events, and hence an attempt to draw back on the syringe after IM<br />

insertion of the needle should be made in order to ensure that the needle is not<br />

in a small vessel. A special product for IV use (NHIG [intravenous]) has been<br />

developed for patients requiring large doses of immunoglobulin. For further<br />

information regarding the use of intravenous immunoglobulins, refer to Criteria<br />

for the clinical use of intravenous immunoglobulin in Australia. 1<br />

Recommendations<br />

Immunoglobulin preparations may be given to susceptible persons, as either<br />

pre-exposure or post-exposure prophylaxis, against specific infections. Normal<br />

pooled immunoglobulin contains sufficiently high antibody concentrations to<br />

be effective against hepatitis A and measles. Both hepatitis A and measles are<br />

notifiable diseases and further instructions about their management and the<br />

need for immunoglobulin can be found in national guidelines (www.health.<br />

gov.au/cdnasongs) and obtained from state/territory public health authorities<br />

(see Appendix 1 Contact details for <strong>Australian</strong>, state and territory government health<br />

authorities and communicable disease control).<br />

<strong>The</strong> duration of effect of NHIG is dose-related. It is estimated that protection is<br />

maintained for 3 to 4 weeks with standard recommended doses of NHIG.<br />

Prevention of hepatitis A<br />

Hepatitis A vaccination (see 4.4 Hepatitis A) is recommended in preference to<br />

NHIG for post-exposure hepatitis A prophylaxis in persons ≥12 months of age<br />

who are immunocompetent.<br />

NHIG can be used when hepatitis A vaccine administration is contraindicated,<br />

in infants


Prevention of measles<br />

Measles vaccination (see 4.9 Measles) within 72 hours of case contact is<br />

recommended in preference to NHIG for post-exposure measles prophylaxis in<br />

many instances (see Table 4.9.2 in 4.9 Measles).<br />

NHIG contains a sufficiently high concentration of antibody against measles<br />

to be able to prevent or ameliorate infection in susceptible persons. NHIG<br />

should be given as soon as possible and within 7 days of exposure. 3 Passive<br />

protection, against measles particularly, may be required if the exposed person<br />

has an underlying immunological disorder (HIV/AIDS, immunosuppressive<br />

therapy), or to control an outbreak of measles among non-immunised persons,<br />

for example, in a childcare centre. <strong>The</strong> use of NHIG should be considered in<br />

HIV-positive persons exposed to a patient with measles.<br />

Immune deficiency<br />

Patients with abnormal antibody production (primary<br />

hypogammaglobulinaemia, multiple myeloma, chronic lymphoblastic<br />

leukaemia) usually receive therapy with the IV preparation of normal human<br />

immunoglobulin (NHIG [intravenous]). However, in some cases, NHIG is given<br />

by IM injection. <strong>The</strong> aim of therapy is to maintain serum IgG levels above 6 g/L.<br />

Some patients may receive the IM (160 mg/mL) preparation subcutaneously. For<br />

further information regarding the use of intravenous immunoglobulins, refer to<br />

Criteria for the clinical use of intravenous immunoglobulin in Australia. 1<br />

Note: Skin tests with NHIG should not be undertaken. <strong>The</strong> intradermal injection<br />

of concentrated immunoglobulin causes a localised area of inflammation, which<br />

can be misinterpreted as a positive allergic reaction. True allergic responses to<br />

NHIG given by IM injection are extremely rare.<br />

5.1.4 Specific immunoglobulins<br />

Specific immunoglobulins are used to protect against specific microbial<br />

agents such as hepatitis B, rabies and varicella-zoster viruses, and tetanus.<br />

Further instructions about the management of these diseases and the need<br />

for immunoglobulin should be obtained from state/territory public health<br />

authorities (see Appendix 1 Contact details for <strong>Australian</strong>, state and territory<br />

government health authorities and communicable disease control), and the national<br />

guidelines for management of disease from rabies and other lyssaviruses,<br />

including <strong>Australian</strong> bat lyssavirus (www.health.gov.au/cdnasongs).<br />

Specific immunoglobulins for botulism and cytomegalovirus (CMV) and<br />

a monoclonal antibody preparation for respiratory syncytial virus (RSV)<br />

are available as described below. Potential interactions, adverse events and<br />

storage requirements for these specific immunoglobulins are similar to those<br />

for NHIG (IM).<br />

PART 5 PASSIVE IMMUNISATION 459<br />

5.1 PASSIVE IMMUNISATION<br />

USING IMMUNOGLOBULIN<br />

PREPARATIONS


Hepatitis B specific immunoglobulin<br />

Hepatitis B specific immunoglobulin (HBIG) is prepared from plasma donated<br />

through routine blood bank collection. Stocks of HBIG are very limited, and use<br />

should be strictly reserved for those who are at high risk, such as babies born<br />

to mothers with chronic hepatitis B infection and non-immune persons who are<br />

exposed through occupational exposure to the blood of unidentified persons, or<br />

to persons who are chronically infected with hepatitis B or whose hepatitis status<br />

cannot be ascertained in time. 4 Requests for HBIG should be directed to the<br />

<strong>Australian</strong> Red Cross Blood Service in your state/territory (see 5.1.1 Availability of<br />

immunoglobulins above).<br />

See 4.5 Hepatitis B, ‘Management of infants born to mothers who are HBsAgpositive’<br />

in 4.5.7 Recommendations and 4.5.11 Public health management of hepatitis B,<br />

for more information.<br />

Rabies specific immunoglobulin<br />

Rabies specific immunoglobulin (HRIG) is prepared from the plasma of<br />

hyperimmunised human donors. HRIG is only administered in persons who<br />

have not received a previous course of rabies vaccine. HRIG is also administered<br />

as part of the post-exposure prophylaxis used following potential <strong>Australian</strong> bat<br />

lyssavirus or other lyssavirus exposures in previously unvaccinated persons. 5<br />

A single dose of HRIG is given to provide localised anti-rabies antibody<br />

protection while the patient responds to the rabies vaccine. It should be given<br />

at the same time as the 1st post-exposure dose of vaccine (day 0). If not given<br />

with the 1st vaccine dose, it may be given up to day 7. From day 8 onwards, an<br />

antibody response to rabies vaccine is presumed to have occurred.<br />

<strong>The</strong> dose of HRIG is based on body mass and should be infiltrated in and<br />

around all wounds, using as much of the calculated HRIG dose as possible.<br />

<strong>The</strong> remainder of the HRIG dose should be administered intramuscularly at a<br />

site away from the injection site of rabies vaccine.<br />

See 4.16 Rabies and other lyssaviruses (including <strong>Australian</strong> bat lyssavirus) for more<br />

information.<br />

Varicella-zoster specific immunoglobulin<br />

Zoster immunoglobulin (ZIG) is highly efficacious, but is often in short supply.<br />

Normal high-titre zoster immunoglobulin is available from the <strong>Australian</strong> Red<br />

Cross Blood Service on a restricted basis for the prevention of varicella in highrisk<br />

subjects who report a significant exposure to varicella or herpes zoster. If ZIG<br />

is unavailable, large doses of NHIG can be given intramuscularly. This does not<br />

necessarily prevent varicella, but it lessens the severity of the disease. ZIG has no<br />

proven use in the treatment of established varicella or zoster infection. ZIG must<br />

be given early in the incubation period (within 96 hours of exposure), but may<br />

have some efficacy if administered out to as late as 10 days post exposure. ZIG<br />

is able to prevent or ameliorate varicella in infants


who are being treated with immunosuppressive therapy, and in pregnant<br />

women. 6,7 Patients suffering from primary or acquired diseases associated with<br />

cellular immune deficiency and those receiving immunosuppressive therapy<br />

should be tested for varicella-zoster antibodies following contact with a person<br />

with confirmed varicella. However, this should not delay ZIG administration,<br />

preferably within 96 hours and up to 10 days after initial exposure. 7<br />

See 4.22 Varicella for more information.<br />

Botulism antitoxin<br />

An equine antitoxin (derived from horses) has long been used in the treatment of<br />

adult botulism, but has not been shown to be effective in infant botulism. 8 Equine<br />

antitoxin is manufactured by pharmaceutical companies such as Chiron. Use in<br />

Australia is governed by the <strong>The</strong>rapeutic Goods Administration’s Special Access<br />

Scheme and physicians wishing to access this product should initially contact<br />

the relevant state/territory health authority (see Appendix 1 Contact details for<br />

<strong>Australian</strong>, state and territory government health authorities and communicable disease<br />

control). Hypersensitivity, presenting as fever, serum sickness or anaphylaxis,<br />

may follow the use of equine antitoxin. Skin testing followed by appropriate<br />

dosing should be administered according to the manufacturer’s instructions.<br />

An intravenous botulinum antitoxin, produced in the United States (BabyBIG; its<br />

sponsor is the Californian Department of Health Services), significantly reduces<br />

the duration of mechanical ventilation and hospitalisation in infant’s with<br />

botulism. 9 This product has been administered to <strong>Australian</strong> children with infant<br />

botulism. 10 It is not currently registered in Australia, but is registered by the<br />

United States Food and Drug Authority. Access to this product should be sought<br />

through the TGA’s Special Access Scheme.<br />

Cytomegalovirus immunoglobulin<br />

Cytomegalovirus (CMV) immunoglobulin is indicated for the prevention of CMV<br />

infection in immunocompromised persons at high risk of severe CMV disease,<br />

such as after bone marrow and renal transplants. 11-13 <strong>The</strong> treatment of established<br />

CMV infection and disease is primarily with antivirals, such as ganciclovir or<br />

vanciclovir, and there is contradictory evidence whether the addition of CMV<br />

immunoglobulin improves outcome. 11,13<br />

<strong>The</strong> product contains no antibacterial agent, and so it must be used immediately<br />

after opening. Any unused portion must be discarded. If the solution has been<br />

frozen, it must not be used. If the use of CMV immunoglobulin is contemplated,<br />

detailed protocols for administration and management of adverse events should<br />

be consulted, in addition to the product information.<br />

• CMV Immunoglobulin-VF (human) – CSL Limited. 55–65 mg/mL<br />

immunoglobulin (mainly IgG) prepared from human plasma with<br />

high levels of antibody to CMV. Single vials contain 1.5 million units<br />

of CMV immunoglobulin activity. Contains maltose.<br />

PART 5 PASSIVE IMMUNISATION 461<br />

5.1 PASSIVE IMMUNISATION<br />

USING IMMUNOGLOBULIN<br />

PREPARATIONS


Respiratory syncytial virus monoclonal antibodies<br />

A humanised mouse monoclonal antibody to respiratory syncytial virus (RSV)<br />

produced by cultured cells, palivizumab, is registered in Australia for prevention<br />

of serious lower respiratory tract disease caused by RSV in children at high<br />

risk of RSV disease. <strong>The</strong>re is no consensus regarding the use of palivizumab<br />

in Australia. 14 This product is given by IM injection each month to children at<br />

high risk of severe RSV disease, during the seasonal period of exposure to RSV.<br />

Palivizumab has been found to reduce the absolute risk of hospitalisation from<br />

about 10% to about 5% for babies born prematurely, for babies with chronic<br />

neonatal lung disease, and also for babies with haemodynamically significant<br />

congenital heart disease, particularly when complicated by large left-to-right<br />

shunts leading to pulmonary hypertension. 14-22 It has not been shown to reduce<br />

the incidence of more severe outcomes, such as the need for ventilation, nor has it<br />

been shown to reduce mortality. 20,21 <strong>The</strong>re are currently a number of clinical trials<br />

assessing a recombinant humanised antibody, motavizumab. 23<br />

• Synagis – Abbott Australia (palivizumab). Supplied in single-use<br />

vials of powder, to be reconstituted with sterile water for injection;<br />

50 mg in 4 mL vial; 100 mg in 10 mL vial.<br />

<strong>The</strong> dose of palivizumab is 15 mg/kg once a month, to be given by IM injection,<br />

preferably in the anterolateral thigh. Where possible, the 1st dose should be<br />

administered before commencement of the RSV season.<br />

Tetanus immunoglobulin<br />

Tetanus immunoglobulin (human) for intramuscular use<br />

Tetanus immunoglobulin (TIG) should be used for passive protection of<br />

persons who have sustained a tetanus-prone wound, where the person has not<br />

previously received 3 or more doses of a tetanus toxoid-containing vaccine or<br />

where there is doubt about their tetanus vaccination status. In persons who have<br />

a humoral immune deficiency, TIG should be provided after a tetanus-prone<br />

injury, regardless of the time since their last dose of tetanus-containing vaccine.<br />

TIG provides immediate protection that lasts for a period of 3 to 4 weeks. 24 For<br />

wounds not categorised as tetanus-prone, such as clean cuts, TIG is unnecessary.<br />

Detailed information on appropriate tetanus prophylaxis measures in wound<br />

management, including use of TIG, are outlined in Table 4.19.1 in 4.19 Tetanus.<br />

<strong>The</strong> recommended dose for TIG is 250 IU, to be given by IM injection as soon as<br />

practicable after the injury. If more than 24 hours have elapsed, 500 IU should be<br />

given. Because of its viscosity, TIG should be given to adults using a 21 gauge<br />

needle. For children, it can be given slowly using a 23 gauge needle. A tetanus<br />

toxoid-containing vaccine should be given at the same time in the opposite<br />

462 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


limb with a separate syringe, and arrangements should be made to complete<br />

the full course of tetanus toxoid-containing vaccinations. Details for accessing<br />

TIG should be obtained from the <strong>Australian</strong> Red Cross Blood Service (see 5.1.1<br />

Availability of immunoglobulins above).<br />

Tetanus immunoglobulin (human) for intravenous use<br />

• Tetanus Immunoglobulin-VF (human, for intravenous use) – CSL<br />

Limited. 55–65 mg/mL immunoglobulin (mainly IgG) prepared<br />

from human plasma containing high levels of antibody to the toxin<br />

of Clostridium tetani. Single vials containing 4000 IU human tetanus<br />

antitoxin. Contains maltose.<br />

Tetanus immunoglobulin for IV use (TIVG) is used in the management of clinical<br />

tetanus. 24 <strong>The</strong> recommended dose is 4000 IU, to be given by slow intravenous<br />

infusion. Detailed protocols for administration of this product and management<br />

of adverse events should be consulted if its use is contemplated. Requests for<br />

TIVG should be directed to the <strong>Australian</strong> Red Cross Blood Service in your state/<br />

territory (see 5.1.1 Availability of immunoglobulins above).<br />

Diphtheria antitoxin<br />

Diphtheria antitoxin is prepared by immunising horses against the toxin<br />

produced by Corynebacterium diphtheriae.<br />

Advice should be sought with respect to diphtheria antitoxin access and dosage,<br />

and special arrangements made if hypersensitivity is suspected; this can be<br />

coordinated through the relevant state/territory health authority (see Appendix 1<br />

Contact details for <strong>Australian</strong>, state and territory government health authorities and<br />

communicable disease control).<br />

5.1.5 Potential interaction with vaccines<br />

Live attenuated viral vaccines<br />

Immunoglobulin preparations can interfere with the response to certain<br />

live attenuated viral vaccines by preventing vaccine virus replication after<br />

administration. <strong>The</strong>refore, administration of live attenuated viral vaccines,<br />

such as measles and varicella vaccines (but not rotavirus, zoster or yellow<br />

fever vaccines), should be deferred, dependent on the clinical status of the<br />

patient, for at least 3 months after the IM administration of NHIG, and for at<br />

least 8 months after the administration of intravenous NHIG. 25 For detailed<br />

information on recommended intervals, see 3.3 Groups with special vaccination<br />

requirements, Table 3.3.6 Recommended intervals between either immunoglobulins or<br />

blood products and MMR, MMRV or varicella vaccination. For the same reason, if<br />

vaccination has occurred, administration of immunoglobulin products should be<br />

PART 5 PASSIVE IMMUNISATION 463<br />

5.1 PASSIVE IMMUNISATION<br />

USING IMMUNOGLOBULIN<br />

PREPARATIONS


deferred if possible until at least 3 weeks after a measles-containing or varicellacontaining<br />

vaccine has been given, unless it is essential that immunoglobulin be<br />

administered. However, Rh (D) immunoglobulin (anti-D) does not interfere with<br />

the antibody response to MMR- or varicella-containing vaccines and the two may<br />

be given at the same time in different sites with separate syringes or at any time<br />

in relation to each other (see 3.3 Groups with special vaccination requirements, Table<br />

3.3.6 Recommended intervals between either immunoglobulins or blood products and<br />

MMR, MMRV or varicella vaccination).<br />

Inactivated vaccines<br />

Inactivated vaccines, such as tetanus, hepatitis B or rabies, may be administered<br />

concurrently with immunoglobulin preparations, or at any time before or after<br />

receipt of immunoglobulin, using separate syringes and separate injection sites.<br />

This usually would occur when there has been actual or possible acute exposure<br />

to one of these infectious agents.<br />

5.1.6 Use in pregnancy<br />

Refer to 3.3 Groups with special vaccination requirements, Table 3.3.1<br />

Recommendations for vaccination in pregnancy for more information.<br />

5.1.7 Contraindications<br />

Hypersensitivity reactions to immunoglobulin preparations occur rarely but<br />

may be more common in patients receiving repeated injections. Intramuscular<br />

immunoglobulins should not be administered to persons who have severe<br />

thrombocytopenia or any coagulation disorder that would contraindicate<br />

intramuscular injections.<br />

5.1.8 Adverse events and precautions<br />

Local tenderness, erythema and muscle stiffness at the site of injection occur very<br />

commonly (in over 10% of recipients) and may persist for several hours after<br />

injection. Systemic adverse events such as mild pyrexia, malaise, drowsiness,<br />

urticaria and angioedema are uncommon, occurring in fewer than 1% of<br />

recipients). Skin lesions, headache, dizziness, nausea, general hypersensitivity<br />

reactions and convulsions may occur rarely.<br />

Anaphylaxis following an injection of NHIG is very rare, but has been reported.<br />

Anaphylaxis is more likely to occur if NHIG for IM use is inadvertently given<br />

intravenously.<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

464 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


APPENDIX 1: CONTACT DETAILS FOR AUSTRALIAN,<br />

STATE AND TERRITORY GOVERNMENT<br />

HEALTH AUTHORITIES AND<br />

COMMUNICABLE DISEASE CONTROL*<br />

<strong>Australian</strong> Government health authorities<br />

<strong>Australian</strong> Government 02 6289 1555<br />

Department of Health<br />

Freecall: 1800 671 811<br />

and Ageing<br />

www.immunise.health.gov.au<br />

<strong>Australian</strong> Childhood<br />

<strong>Immunisation</strong> Register<br />

enquiries (ACIR) †<br />

1800 653 809<br />

ACIR email: acir@humanservices.gov.au<br />

ACIR Internet site: www.humanservices.gov.au/<br />

customer/services/medicare/australian-childhoodimmunisation-register<br />

State and territory government health authorities<br />

<strong>Australian</strong> Capital 02 6205 2300<br />

Territory<br />

<strong>Immunisation</strong> Enquiry Line<br />

New South Wales 1300 066 055<br />

(to connect to your local Public Health Unit)<br />

Northern Territory 08 8922 8044<br />

Centre for Disease Control<br />

Queensland 13 HEALTH (13 4325 84)<br />

Contact your local Public Health Unit, details at<br />

www.health.qld.gov.au/cdcg/contacts.asp<br />

South Australia 1300 232 272 (8.30 am to 5.00 pm)<br />

Email: CDCB@health.sa.gov.au<br />

www.sahealth.sa.gov.au<br />

Tasmania 03 6222 7666 or 1800 671 738<br />

Victoria 1300 882 008<br />

Email: immunisation@health.vic.gov.au<br />

www.health.vic.gov.au/immunisation<br />

Western Australia 08 9388 4868<br />

08 9328 0553 (after hours Infectious Diseases<br />

Emergency)<br />

Email: cdc@health.wa.gov.au<br />

Contact details for communicable disease control<br />

<strong>Australian</strong> Capital 24-hour Communicable Disease Control Section:<br />

Territory<br />

02 6205 2155<br />

APPENDIX 1 465<br />

APPENDIX 1


New South Wales 1300 066 055<br />

(for connection to Public Health Unit)<br />

Northern Territory 8.30 am to 5.00 pm: 08 8922 8044 Centre for Disease<br />

Control<br />

(After hours Royal Darwin Hospital 08 8922 8888 for<br />

CDC on-call doctor)<br />

Queensland Contact your local Public/Population Health Unit,<br />

phone number listed in the White Pages<br />

South Australia 24 hour general enquiries line: 1300 232 272<br />

Tasmania 24 hour hotline: 1800 671 738<br />

Victoria 24 hour contact number: 1300 651 160<br />

Western Australia Perth Metropolitan area:<br />

08 9388 4852<br />

After hours/Emergency: 08 9328 0533<br />

Outside Perth Metropolitan area:<br />

Contact regional Population Health Unit<br />

* See also state/territory and <strong>The</strong>rapeutic Goods Administration (TGA) contact details for<br />

reporting AEFI in Table 2.3.3 Contact information for notification of adverse events following<br />

immunisation in 2.3.2 Adverse events following immunisation.<br />

† For more information on other registers, see 2.3.4 <strong>Immunisation</strong> registers.<br />

466 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


APPENDIX 2: LITERATURE SEARCH STRATEGY FOR<br />

THE 10TH EDITION OF THE HANDBOOK<br />

For each <strong>Handbook</strong> chapter, broad literature searches were conducted for the years<br />

since the last <strong>Handbook</strong> searches were performed, using up to 24 databases, listed<br />

in Table A2.1. <strong>The</strong> purpose of these searches was to ensure that NCIRS technical<br />

writers and ATAGI members had access to all relevant information from the<br />

latest medical literature to allow identification of important issues related to<br />

the updating of all <strong>Handbook</strong> chapters. In addition, since writing of the 9th<br />

edition of the <strong>Handbook</strong>, Selected Dissemination Information (SDI) searches were<br />

established to enable the ongoing collection of new relevant items on the search<br />

topics. This process used the same search strategies as previously described,<br />

which allowed consideration and inclusion of papers published since publication<br />

of the 9th edition <strong>Handbook</strong>.<br />

Table A2.1: Electronic databases searched for the <strong>10th</strong> edition<br />

Electronic database Time period<br />

MEDLINE 2006–2011<br />

Cochrane Library – including Cochrane Database of Systematic 2006–2011<br />

Reviews, Database of Abstracts of Reviews of Effects, the<br />

Cochrane Central Register of Controlled Trials (CENTRAL),<br />

Methods Studies, the Health Technology Assessment Database,<br />

the NHS Economic Evaluation Database<br />

Cumulated Index Nursing & Allied Health Literature (CINAHL) 2006–2011<br />

(when required)<br />

Clinical Evidence 2006–2011<br />

EMBASE 2006–2011<br />

<strong>Australian</strong> focused Informit databases (AMI, APA-FT, APAIS, 2006–2011<br />

APAIS - Health, ATSIhealth, Ausport Med, CINCH - Health,<br />

DRUG, Health and Society, HIVA, Health Collection, Indigenous<br />

Collection, RURAL, SAGE)<br />

Searches were conducted using the electronic databases detailed in Table A2.1,<br />

with the search period from 2006 to October 2011, in order to retrieve items<br />

published since the searches completed for the 9th edition of the <strong>Handbook</strong>. <strong>The</strong><br />

scope of the searches was broad, to ensure maximum retrieval and minimise<br />

the exclusion of items of interest. Previous <strong>Handbook</strong> searches were examined to<br />

determine the scope required for the new searches, and similar search strategies<br />

APPENDIX 2 467<br />

APPENDIX 2


were employed to ensure consistency of information retrieval, taking into<br />

account new terms added to the databases.<br />

Various search methods were tested, including ‘explode’ and ‘focus’ options.<br />

‘Exploded’ terms retrieve citations containing the term being searched and all<br />

the narrower related terms in the database. ‘Focus’ searches retrieve citations<br />

that have the search term as the major focus of the item. In the trial searches,<br />

some items of interest were missed using the ‘focus’ method, thus ‘exploded’<br />

searches were utilised. All subheadings assigned to the subject headings were<br />

generally included. In general, the search strategy consisted of the disease topic<br />

and relevant vaccine terms, used in combination with the terms immunisation/<br />

immunisation programs. Boolean operators AND, OR and NOT were used as<br />

appropriate. To ensure relevant and accurate retrieval, thesaurus terms (the<br />

controlled vocabulary terms used in the database) were used whenever possible.<br />

Keyword searching was used in the absence of an appropriate thesaurus term or<br />

if the database did not have thesaurus terms. To facilitate relevant retrieval and<br />

to limit what, in some instances, are very large search result sets, the following<br />

limits were applied to the disease topic searches:<br />

• Publication year – searches were generally limited to items published from<br />

2006–2011.<br />

• Language – searches were limited to items in English.<br />

• Human – items discussing only animals were removed.<br />

• In vitro – items discussing only in vitro studies were removed.<br />

• Abstracts – search results restricted to items containing abstracts.<br />

<strong>The</strong> search limits were slightly modified for some of the searches. For example,<br />

the <strong>Australian</strong>-specific searches did not have search results limited to abstract<br />

only, to ensure that all <strong>Australian</strong> items were retrieved.<br />

<strong>The</strong> ATAGI and NCIRS technical writers also identified, where possible, focused<br />

clinical questions for each of the <strong>Handbook</strong> chapters, in advance of conducting<br />

literature searches. Specific searches were conducted for these questions, both<br />

using the databases and time periods above, but also using additional databases,<br />

longer time periods and other strategies, such as clinical trial registries and<br />

handsearching, as necessary.<br />

468 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


APPENDIX 3: COMPONENTS OF VACCINES USED IN THE<br />

NATIONAL IMMUNISATION PROGRAM<br />

Please note that vaccine manufacture is subject to ongoing refinement and<br />

change. <strong>The</strong>refore, the information in Table A3.1 may change. This information<br />

was current as of mid-2012 and has been sourced from the product information<br />

(PI) of each vaccine listed. <strong>The</strong> <strong>The</strong>rapeutic Goods Administration provides the<br />

most current versions of the PI and Consumer Medicines Information (CMI)<br />

documents for vaccines (and other medicines) on its website at www.tga.gov.au.<br />

For vaccines not listed in the National <strong>Immunisation</strong> Program, please refer<br />

to individual product information leaflet as supplied with the vaccine, or the<br />

<strong>Handbook</strong> chapter pertinent to that vaccine.<br />

None of the vaccines listed on the National <strong>Immunisation</strong> Program contains<br />

thiomersal.<br />

Table A3.1: Components of vaccines used in the National <strong>Immunisation</strong> Program<br />

Vaccine<br />

component*<br />

Vaccine brand † Antigen<br />

Albumin/serum Avaxim Hepatitis A (HAV)<br />

ProQuad Measles-mumps-rubella-varicella (MMRV)<br />

Quadracel Diphtheria-tetanus-acellular pertussisinactivated<br />

poliomyelitis (DTPa-IPV)<br />

Vaqta Hepatitis A (HAV) paediatric/adolescent<br />

Varilrix Varicella (VV)<br />

Varivax Refrigerated Varicella (VV)<br />

Aluminium Avaxim Hepatitis A (HAV)<br />

hydroxide Cervarix Human papillomavirus (HPV)<br />

Engerix-B Hepatitis B (HBV) adult and paediatric<br />

Havrix Junior Hepatitis A (HAV) paediatric<br />

H-B-Vax II Hepatitis B (HBV) adult and paediatric<br />

Infanrix IPV Diphtheria-tetanus-acellular pertussisinactivated<br />

poliomyelitis (DTPa-IPV)<br />

Menjugate Syringe Serogroup C meningococcal conjugate<br />

(MenCCV)<br />

NeisVac-C Serogroup C meningococcal conjugate<br />

(MenCCV)<br />

Vaqta Hepatitis A (HAV) paediatric/adolescent<br />

APPENDIX 3 469<br />

APPENDIX 3


Vaccine<br />

component*<br />

Aluminium<br />

hydroxide/<br />

phosphate<br />

Aluminium<br />

phosphate<br />

Borax/sodium<br />

borate<br />

Vaccine brand † Antigen<br />

Boostrix Diphtheria-tetanus-acellular pertussis<br />

(dTpa) reduced antigen<br />

Gardasil Human papillomavirus (HPV)<br />

Infanrix hexa Diphtheria-tetanus-acellular pertussishepatitis<br />

B-inactivated poliomyelitis-<br />

Haemophilus influenzae type b<br />

(DTPa-hepB-IPV-Hib)<br />

Adacel Diphtheria-tetanus-acellular pertussis<br />

(dTpa) reduced antigen<br />

Meningitec Serogroup C meningococcal conjugate<br />

(MenCCV)<br />

Prevenar 13 13-valent pneumococcal conjugate<br />

(13vPCV)<br />

Quadracel Diphtheria-tetanus-acellular pertussisinactivated<br />

poliomyelitis (DTPa-IPV)<br />

Gardasil Human papillomavirus (HPV)<br />

Vaqta Hepatitis A (HAV) paediatric/adolescent<br />

Egg protein All influenza vaccines<br />

Agrippal Influenza<br />

Fluarix Influenza<br />

Fluvax Influenza<br />

Influvac Influenza<br />

Vaxigrip Influenza<br />

Formaldehyde Adacel Diphtheria-tetanus-acellular pertussis<br />

(dTpa) reduced antigen<br />

Agrippal Influenza<br />

Avaxim Hepatitis A (HAV)<br />

Boostrix Diphtheria-tetanus-acellular pertussis<br />

(dTpa) reduced antigen<br />

Fluarix Influenza<br />

Havrix Junior Hepatitis A (HAV) paediatric<br />

Infanrix hexa Diphtheria-tetanus-acellular pertussishepatitis<br />

B-inactivated poliomyelitis-<br />

Haemophilus influenzae type b (DTPa-hepB-<br />

IPV-Hib)<br />

Infanrix IPV Diphtheria-tetanus-acellular pertussisinactivated<br />

poliomyelitis (DTPa-IPV)<br />

Influvac Influenza<br />

Quadracel Diphtheria-tetanus-acellular pertussisinactivated<br />

poliomyelitis (DTPa-IPV)<br />

Vaqta Hepatitis A (HAV) paediatric/adolescent<br />

Vaxigrip Influenza<br />

470 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Vaccine<br />

component*<br />

Vaccine brand † Antigen<br />

Gelatin ProQuad Measles-mumps-rubella-varicella (MMRV)<br />

Varivax Refrigerated Varicella (VV)<br />

Gentamicin Fluarix Influenza<br />

Influvac Influenza<br />

Glutaraldehyde Adacel Diphtheria-tetanus-acellular pertussis<br />

(dTpa) reduced antigen<br />

Quadracel Diphtheria-tetanus-acellular pertussisinactivated<br />

poliomyelitis (DTPa-IPV)<br />

Kanamycin Agrippal Influenza<br />

Mannitol Priorix Measles-mumps-rubella (MMR)<br />

Priorix-tetra Measles-mumps-rubella-varicella (MMRV)<br />

Monosodium ProQuad Measles-mumps-rubella-varicella (MMRV)<br />

glutamate (MSG)<br />

Varivax Refrigerated Varicella (VV)<br />

Neomycin Agrippal Influenza<br />

Avaxim Hepatitis A (HAV)<br />

Fluvax Influenza<br />

Havrix Junior Hepatitis A (HAV) paediatric<br />

Infanrix hexa Diphtheria-tetanus-acellular pertussishepatitis<br />

B-inactivated poliomyelitis-<br />

Haemophilus influenzae type b (DTPa-hepB-<br />

IPV-Hib)<br />

Infanrix IPV Diphtheria-tetanus-acellular pertussisinactivated<br />

poliomyelitis (DTPa-IPV)<br />

Priorix Measles-mumps-rubella (MMR)<br />

Priorix-tetra Measles-mumps-rubella-varicella (MMRV)<br />

ProQuad Measles-mumps-rubella-varicella (MMRV)<br />

Quadracel Diphtheria-tetanus-acellular pertussisinactivated<br />

poliomyelitis (DTPa-IPV)<br />

Vaqta Hepatitis A (HAV) paediatric/adolescent<br />

Varilrix Varicella (VV)<br />

Varivax Refrigerated Varivax Refrigerated<br />

Vaxigrip Influenza<br />

Phenol Pneumovax 23 23-valent pneumococcal polysaccharide<br />

(23vPPV)<br />

Phenoxyethanol Adacel Diphtheria-tetanus-acellular pertussis<br />

(dTpa) reduced antigen<br />

Avaxim Hepatitis A (HAV)<br />

Quadracel Diphtheria-tetanus-acellular pertussisinactivated<br />

poliomyelitis (DTPa-IPV)<br />

APPENDIX 3 471<br />

APPENDIX 3


Vaccine<br />

component*<br />

Vaccine brand † Antigen<br />

Polymyxin Fluvax Influenza<br />

Infanrix hexa Diphtheria-tetanus-acellular pertussishepatitis<br />

B-inactivated poliomyelitis-<br />

Haemophilus influenzae type b (DTPa-hepB-<br />

IPV-Hib)<br />

Infanrix IPV Diphtheria-tetanus-acellular pertussisinactivated<br />

poliomyelitis (DTPa-IPV)<br />

Quadracel Diphtheria-tetanus-acellular pertussisinactivated<br />

poliomyelitis (DTPa-IPV)<br />

Polysorbate or Agrippal Influenza<br />

sorbitol Boostrix Diphtheria-tetanus-acellular pertussis<br />

(dTpa) reduced antigen<br />

Fluarix Influenza<br />

Gardasil Human papillomavirus (HPV)<br />

Havrix Junior Hepatitis A (HAV) paediatric<br />

Infanrix hexa Diphtheria-tetanus-acellular pertussishepatitis<br />

B-inactivated poliomyelitis-<br />

Haemophilus influenzae type b (DTPa-hepB-<br />

IPV-Hib)<br />

Infanrix IPV Diphtheria-tetanus-acellular pertussisinactivated<br />

poliomyelitis (DTPa-IPV)<br />

Influvac Influenza<br />

Prevenar 13 13-valent pneumococcal conjugate<br />

(13vPCV)<br />

Priorix Measles-mumps-rubella (MMR)<br />

Priorix-tetra Measles, mumps, rubella and varicella<br />

(MMRV)<br />

ProQuad Measles-mumps-rubella-varicella (MMRV)<br />

Quadracel Diphtheria-tetanus-acellular pertussisinactivated<br />

poliomyelitis (DTPa-IPV)<br />

RotaTeq Rotavirus<br />

Yeast Engerix-B Hepatitis B (HBV) adult and paediatric<br />

Gardasil Human papillomavirus (HPV)<br />

H-B-Vax II Hepatitis B (HBV) adult and paediatric<br />

Infanrix hexa Diphtheria-tetanus-acellular pertussishepatitis<br />

B-inactivated poliomyelitis-<br />

Haemophilus influenzae type b (DTPa-hepB-<br />

IPV-Hib)<br />

* If the person to be vaccinated has had an anaphylactic reaction to any of the vaccine components,<br />

administration of that vaccine may be contraindicated. Specialist advice should be sought to<br />

identify the component and to review if the person can be vaccinated in future.<br />

† Please also refer to Appendix 4 Commonly asked questions about vaccination for more specific<br />

information about these various constituents.<br />

472 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


APPENDIX 4: COMMONLY ASKED QUESTIONS<br />

ABOUT VACCINATION<br />

This appendix contains information for providers to refer to when responding<br />

to questions and concerns about immunisation. It covers general questions on<br />

adult and childhood vaccination, including contraindications and precautions.<br />

In addition, a discussion on some of the more recent concerns about vaccination<br />

is included, covering issues relating to vaccine safety, vaccine content,<br />

immunisation as a possible cause of some illnesses of uncertain origin, and the<br />

need for vaccination.<br />

This appendix is divided into six sections:<br />

• General questions<br />

• Questions about contraindication and precautions<br />

• Questions about vaccine safety<br />

• Questions about vaccine content<br />

• Questions about the need for immunisation<br />

• Further information about vaccination<br />

A.4.1 General questions<br />

How does vaccination work?<br />

When a healthy person becomes infected with a virus or bacteria (also known<br />

as a pathogen), for example, the measles virus, the body recognises the virus as<br />

an invader, produces antibodies that eventually destroy the virus, and recovery<br />

occurs. If contact with the measles virus occurs again in the future, the body’s<br />

immune system ‘remembers’ the measles virus and produces an increase in<br />

antibodies to destroy this pathogen.<br />

Vaccination is the process that is used to stimulate the body’s immune system<br />

in the same way as the real pathogen or disease would, but without causing the<br />

symptoms of the disease. Most vaccines provide the body with ‘memory’ so<br />

that an individual does not get the disease if exposed to it (see 1.5 Fundamentals<br />

of immunisation).<br />

Vaccination conveys immunity to diseases by a process called active immunity,<br />

which can be achieved by administration of either inactivated (i.e. not live) or<br />

live attenuated pathogens or their products. Live vaccines are attenuated, or<br />

weakened, by growing the organism through serial culturing (or passaging)<br />

steps in various tissue culture media. Inactivation is usually done using<br />

heat or formalin (sometimes both). Inactivated vaccines may include the<br />

whole pathogen (such as oral cholera vaccine), the toxin produced by the<br />

pathogen (such as tetanus and diphtheria vaccines), or specific antigens (such<br />

as Haemophilus influenzae type b [Hib], meningococcal and pneumococcal<br />

APPENDIX 4 473<br />

APPENDIX 4


vaccines). In some cases, the antigen is conjugated (i.e. chemically linked) with<br />

proteins to facilitate the immune response. Inactivated viral vaccines may<br />

include whole viruses (such as inactivated poliomyelitis vaccine [IPV] and<br />

hepatitis A vaccines) or specific antigens (such as influenza and hepatitis B<br />

vaccines). Live attenuated viral vaccines include measles-mumps-rubella<br />

(MMR), varicella and yellow fever vaccines.<br />

Immunity can also be acquired passively by the administration of<br />

immunoglobulins, which are the same as antibodies (see 1.5 Fundamentals of<br />

immunisation). Such immunity is immediate and is dose-related and transient.<br />

For example, measles or hepatitis B immunoglobulin can be used promptly after<br />

exposure in an unimmunised person to help reduce the chance of getting measles<br />

or hepatitis B disease from the exposure.<br />

What is the correct site for vaccination?<br />

<strong>The</strong> top, outer part of the thigh (the vastus lateralis muscle) is the recommended<br />

site for injections for infants


When should preterm infants be vaccinated?<br />

Babies born at 65 years, regardless of the presence or absence of chronic illness, reduces<br />

mortality during the winter period in this age group (see 4.7 Influenza). <strong>The</strong><br />

healthy elderly should also receive the 23-valent pneumococcal polysaccharide<br />

vaccine (see 4.13 Pneumococcal disease).<br />

Should adults receive pertussis (whooping cough) vaccine boosters?<br />

Yes. Two brands of acellular pertussis vaccines, both combined with tetanus and<br />

diphtheria antigens, are now available for adolescents and adults. dTpa vaccines<br />

are recommended in Australia for booster vaccination of individuals ≥10 years of<br />

age who have previously had a primary course of diphtheria-tetanus-pertussis<br />

vaccine. dTpa vaccines have a lower content of diphtheria and pertussis antigens<br />

than DTPa formulations for young children.<br />

A recent study showed that adults can be protected against pertussis after a<br />

single dose of dTpa. No recommendations about the need for further boosters<br />

using reduced antigen content formulation dTpa have been made at this time.<br />

A single dose of dTpa is recommended for the following groups, unless<br />

contraindicated or if they have already received a previous dose of dTpa in the<br />

last 10 years (or 5 years when specifically indicated, see 4.12 Pertussis):<br />

• adults working with young children; vaccination is especially recommended<br />

for those working in early childhood education and care<br />

• all healthcare workers<br />

APPENDIX 4 475<br />

APPENDIX 4


• adults planning a pregnancy, or both parents as soon as possible after<br />

delivery of an infant (preferably before hospital discharge)<br />

Pregnant women can also be vaccinated during the last trimester of<br />

pregnancy, as an alternative to getting vaccinated straight after delivery. This<br />

allows for antibodies to be transferred to the infant during the pregnancy<br />

– this particularly helps to protect the infant against pertussis. Other adult<br />

household members, grandparents and carers of young children should<br />

also be vaccinated. This recommendation is based on evidence from several<br />

studies of infant pertussis cases, in which family members, particularly<br />

parents, were identified as the source of infection in more than 50% of cases,<br />

and were the presumed source in a higher proportion.<br />

• any adult expressing an interest in receiving a booster dose of dTpa.<br />

Adults ≥50 years of age who have not previously received dTpa vaccine should<br />

also be offered vaccination (see 4.2 Diphtheria, 4.12 Pertussis or 4.19 Tetanus in this<br />

<strong>Handbook</strong>).<br />

Contraindications to the reduced antigen content formulation dTpa are discussed<br />

in 4.2 Diphtheria, 4.12 Pertussis and 4.19 Tetanus in this <strong>Handbook</strong> and include<br />

previous anaphylactic reaction to any vaccine component.<br />

If the patient has never received a primary course of dT, see 4.2 Diphtheria,<br />

4.12 Pertussis or 4.19 Tetanus in this <strong>Handbook</strong>.<br />

A person wants to receive his/her vaccines separately. Why can’t they do this?<br />

<strong>The</strong>re is no scientific evidence or data to suggest that there are any benefits<br />

in receiving vaccines such as MMR as separate monovalent vaccines. Using<br />

the example of MMR vaccine, there is no individual mumps, measles or<br />

rubella vaccine approved for use in Australia. If these vaccines were to be<br />

administered individually, it would require three separate vaccines, which would<br />

unnecessarily increase discomfort for the child. In addition, if these monovalent<br />

vaccines were not given on the same day, they would need to be spaced 1 month<br />

or more apart, which would increase the risk of that person being exposed to<br />

serious vaccine-preventable diseases. A policy of providing separate vaccines<br />

would cause some people to not receive the entire course. Combination vaccines<br />

can offer a reduced amount of vaccine preparation to be injected overall,<br />

compared to three individual vaccine doses.<br />

Is vaccination compulsory? What happens if children do not get vaccinated?<br />

Vaccination is not compulsory in Australia.<br />

Eligibility for the Family Tax Benefit Part A supplement will require either that<br />

children are assessed as fully immunised or that the parent has obtained an<br />

appropriate medical or philosophical exemption. This replaced the Maternity<br />

<strong>Immunisation</strong> Allowance on 1 July 2012.<br />

476 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


If a parent decides not to have a child vaccinated and, if cases of certain vaccinepreventable<br />

diseases occur at that child’s day-care centre or school, the parent<br />

may, in some circumstances, be required to keep the unvaccinated child at home<br />

until the incubation period for that particular disease has passed or no further<br />

cases have occurred in that setting.<br />

A4.2 Questions about contraindications and precautions<br />

If a person has any concerns about whether to proceed with vaccination, they<br />

should be provided with appropriate information and encouraged to obtain<br />

expert advice from their usual immunisation provider or an immunisation<br />

specialist, if necessary. See Appendix 1 Contact details for <strong>Australian</strong>, state<br />

and territory government health authorities and communicable disease control for<br />

contact details.<br />

What are the absolute contraindications to childhood vaccination?<br />

True contraindications to vaccines are extremely rare (see relevant chapters),<br />

and include only anaphylaxis to any of the particular vaccine’s components,<br />

and anaphylaxis following a previous dose of that vaccine. Follow-up specialist<br />

medical advice should always be sought if any severe reaction or anaphylaxis has<br />

occurred following the administration of any vaccine(s).<br />

Note: Anaphylaxis following ingestion of eggs does not contraindicate MMR<br />

vaccine, as the vaccine viruses are not grown in eggs and the vaccine does not<br />

contain any egg protein (see 4.9 Measles). Many persons who have a history of a<br />

severe allergic reaction to eggs can also be vaccinated with influenza vaccine (see<br />

4.7 Influenza).<br />

Can someone who has had whooping cough (pertussis) still be vaccinated?<br />

Vaccination with pertussis vaccine in children, adolescents or adults who have<br />

had laboratory-confirmed pertussis infection is safe and is necessary, as natural<br />

immunity does not confer life-long protection. In particular, incompletely<br />

vaccinated infants


Should a person with an intercurrent illness be vaccinated?<br />

A child or adult with a minor illness (without systemic illness and with a<br />

temperature


Should vaccines be given to persons who have problems with their<br />

immune systems?<br />

Persons who are immunocompromised (from either a disease or medical<br />

treatment) should generally not be given live viral vaccines such as MMR,<br />

MMRV, varicella, zoster or rotavirus vaccines (see 4.9 Measles, 4.22 Varicella,<br />

4.24 Zoster and 4.17 Rotavirus).<br />

HIV-infected persons may be given MMR, varicella and zoster vaccines,<br />

provided they do not have severe immunocompromise (see 3.3 Groups with special<br />

vaccination requirements and Table 3.3.4 Categories of immunocompromise in HIVinfected<br />

persons, based on age-specific CD4 + counts and percentage of total lymphocytes).<br />

<strong>The</strong> close contacts of persons who are immunocompromised can be given live<br />

viral vaccines, except oral polio vaccine, which is no longer used in Australia.<br />

<strong>The</strong> rash seen in a small percentage of MMR vaccine recipients, usually between<br />

5 and 12 days after vaccination, is not infectious. Non-immune household<br />

contacts of persons who are immunocompromised should receive varicella<br />

vaccine. <strong>The</strong>re is an almost negligible risk of transmitting varicella vaccine virus<br />

from a vaccine-related vesicular rash to contacts. However, vaccine-related<br />

rash occurs in 3 to 5% of vaccinated persons, either locally at the injection site<br />

or generalised, with a median of only 25 lesions. This small infection risk of the<br />

less virulent attenuated vaccine strain is far outweighed by the high risk of nonimmune<br />

contacts catching wild varicella infection and transmitting the virus<br />

to the immunocompromised household member via respiratory droplets or<br />

from the large number of skin lesions that occur with wild varicella infection (a<br />

median of 300 to 500 lesions).<br />

Live viral vaccines can be given to persons with leukaemia and other<br />

malignancies at least 3 months after they have completed chemotherapy,<br />

provided there are no concerns about their immune status. Such measures would<br />

normally be carried out under the supervision of the person’s oncologist (see<br />

3.3.3 Vaccination of immunocompromised persons).<br />

What vaccines should someone with HIV infection receive?<br />

Persons with HIV (human immunodeficiency virus) infection, especially<br />

children, should have all routine inactivated vaccines on the National<br />

<strong>Immunisation</strong> Program schedule. Varicella vaccine is contraindicated in<br />

persons with HIV who are significantly immunocompromised, as it can<br />

cause disseminated varicella infection. However, it may be considered for<br />

asymptomatic or mildly symptomatic HIV-infected children, after weighing<br />

up the potential risks and benefits. This should be discussed with the child’s<br />

specialist. MMR vaccine can be given to children with HIV, depending on their<br />

CD4 + counts (see ‘Should vaccines be given to persons who have problems<br />

with their immune systems?’ above). Persons with HIV infection should also<br />

be vaccinated against pneumococcal disease (see 4.13 Pneumococcal disease).<br />

Influenza vaccine is also recommended for HIV-infected persons. <strong>The</strong>y should<br />

APPENDIX 4 479<br />

APPENDIX 4


not be given BCG, due to the risk of disseminated infection. More detailed<br />

information on the use of vaccines in persons with HIV is included in 3.3.3<br />

Vaccination of immunocompromised persons.<br />

Should chronically ill persons be vaccinated?<br />

In general, persons with chronic diseases should be vaccinated as a matter of<br />

priority, because they are often more at risk from complications from vaccinepreventable<br />

diseases. Annual influenza vaccine is highly recommended for<br />

chronically ill persons and their household contacts.<br />

Care is needed with the use of live attenuated viral vaccines in situations<br />

where the person’s illness, or its treatment, may result in impaired immunity.<br />

Advice may need to be sought on these patients to clarify the safety of live<br />

viral vaccine doses.<br />

Should children or household contacts be vaccinated while the child’s<br />

mother is pregnant?<br />

<strong>The</strong>re is no problem with giving routine vaccinations to a child, or others, living<br />

in the same household with a pregnant woman. MMR vaccine viruses are not<br />

transmissible. Administration of varicella vaccine to household contacts of<br />

non-immune pregnant women is safe. Transmission of varicella vaccine virus<br />

is very rare. <strong>The</strong>re is an almost negligible risk of transmitting varicella vaccine<br />

virus from a vaccine-related vesicular rash to contacts. However, vaccine-related<br />

rash occurs in 3 to 5% of vaccinated persons, either locally at the injection site or<br />

generalised, with a median of only 25 lesions. Furthermore, vaccinating the child<br />

of a pregnant mother will reduce the risk of her being infected by her offspring<br />

with the more virulent wild virus strain if she is not immune (see 3.3.2 Vaccination<br />

of women who are planning pregnancy, pregnant or breastfeeding, and preterm infants).<br />

Should persons with allergies be vaccinated? What precautions are required for<br />

atopic or egg-sensitive children or adults?<br />

Depending on the allergy identified, there often may not be a contraindication<br />

to vaccination. Specialist medical advice should always be sought in order to<br />

determine which vaccinations can be safely given. For example, a history of an<br />

allergy to antibiotics most commonly relates to β-lactam, or related antibiotics,<br />

and is not a contraindication to vaccines that contain neomycin, polymyxin B<br />

or gentamicin. Previous reactions to neomycin that only involved the skin<br />

are not considered a risk factor for a severe allergic reaction or anaphylaxis to<br />

vaccines manufactured with neomycin, since there are only trace amounts of<br />

this antibiotic in the final product (see 3.3.1 Vaccination of persons who have had an<br />

adverse event following immunisation).<br />

For other allergies, see Appendix 3, Components of vaccines used in the National<br />

<strong>Immunisation</strong> Program, and the relevant vaccine product information (PI) enclosed<br />

in the vaccine package. Unless the person being vaccinated has an allergy to a<br />

specific constituent of a vaccine (or has another contraindication), there is no<br />

480 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


eason not to vaccinate. Asthma, eczema and hay fever are not contraindications<br />

to any vaccine, unless the child/adult is receiving high-dose oral steroid therapy.<br />

Persons with egg allergies can receive MMR vaccines because the measles and<br />

mumps components of MMR vaccine do not contain sufficient amounts of egg<br />

ovalbumin to contraindicate MMR vaccination of people with egg allergy (even<br />

anaphylaxis) (see 3.3.1 Vaccination of persons who have had an adverse event following<br />

immunisation and 4.9 Measles). A simple dislike of eggs, or having diarrhoea or<br />

stomach pains after eating eggs, are not reasons to avoid MMR vaccination, and<br />

no special precautions are required in these circumstances. <strong>The</strong>se persons can<br />

also have all other routine vaccines without special precautions.<br />

A history of anaphylaxis or allergy to egg had previously been considered an<br />

absolute contraindication to influenza vaccination, but there have now been<br />

a number of studies indicating that the majority of such persons can be safely<br />

vaccinated. 1,2 Given that there is still a small risk of anaphylaxis, it is essential<br />

that such persons are vaccinated in facilities with staff able to recognise and treat<br />

anaphylaxis. (See also 4.7 Influenza.)<br />

Yellow fever, Q fever and one of the available rabies vaccines contain a higher<br />

amount of egg albumin than is present in the currently available influenza<br />

vaccines. Persons with egg allergy requiring vaccination with either yellow fever,<br />

rabies or Q fever vaccines, should seek specialist immunisation advice from their<br />

state or territory health department. See also relevant chapters of this <strong>Handbook</strong>.<br />

Families with questions about allergies and vaccines are encouraged to discuss<br />

this with their immunisation service provider and, where necessary, seek<br />

referral to an immunologist to have any questions promptly answered to avoid<br />

unnecessary delays of vaccine doses or referral to a specialist immunisation<br />

clinic. Information on specialist immunisation clinics is available from your local<br />

state or territory health department. (See Appendix 1 for further details.)<br />

A4.3 Questions about vaccine safety<br />

Some people have concerns about immunisation. <strong>The</strong>se mostly relate to whether<br />

the vaccine is safe and whether vaccines weaken the immune system. Providers<br />

should always listen to and acknowledge people’s concerns. Providers should<br />

discuss the risks and benefits of immunisation with parents/carers honestly and<br />

in a non-defensive manner. Parents/carers and adult vaccine recipients should<br />

receive accurate information on the risks from vaccine-preventable diseases and<br />

information about vaccine side effects and adverse events (see table Comparison<br />

of the effects of diseases and the side effects of NIP vaccines inside the back cover of<br />

this <strong>Handbook</strong>). <strong>The</strong> following section responds to some concerns raised about the<br />

safety of immunisation, and examines the scientific evidence in order to assist<br />

providers and parents in making an informed choice about the risks and benefits<br />

of vaccination.<br />

APPENDIX 4 481<br />

APPENDIX 4


How safe are vaccines?<br />

Before vaccines are made available for general use they are tested for safety<br />

and efficacy in clinical trials and then in large trials, otherwise known as phase<br />

II (2) and III (3) trials. All vaccines marketed in Australia are manufactured<br />

according to strict safety guidelines and are evaluated by the <strong>The</strong>rapeutic Goods<br />

Administration, to ensure they are efficacious and are of adequate quality and<br />

safety, before marketing approval is granted.<br />

After vaccines are introduced into vaccination schedules, they are subjected to<br />

continuing surveillance of efficacy and safety through trials and post-marketing<br />

surveillance. In Australia, there are regional and national surveillance systems<br />

actively seeking any adverse events following immunisation. This is necessary,<br />

as sometimes unexpected side effects occur after vaccines are registered for use.<br />

<strong>Australian</strong> reports on adverse events that occur following immunisation are<br />

published on a 6-monthly basis in the journal Communicable Diseases Intelligence<br />

(www.health.gov.au/internet/main/publishing.nsf/content/cda-pubs-cdicdiintro.htm).<br />

Can too many vaccines overload or suppress the natural immune system?<br />

No. Although the increase in the number of vaccines and vaccine doses given<br />

to children has led to concerns about the possibility of adverse effects of the<br />

aggregate vaccine exposure, especially on the developing immune system, there<br />

is not a problem. In day-to-day life, all children and adults confront enormous<br />

numbers of antigens, and the immune system responds to each of these in<br />

various ways to protect the body. Studies of the diversity of antigen receptors<br />

indicate that the immune system can respond to an extremely large number of<br />

antigens. In addition, the number of antigens received by children during routine<br />

childhood vaccination has actually decreased compared with several decades<br />

ago. This has occurred in spite of the increase in the total number of vaccines<br />

given, and can be accounted for by the removal of two vaccines – smallpox<br />

vaccine (which contained about 200 different proteins), and whole-cell pertussis<br />

vaccine (about 3000 distinct antigenic components) from routine vaccination<br />

schedules. In comparison, the acellular pertussis vaccine currently used in<br />

Australia has only 3 to 5 pertussis antigens. 3<br />

Do vaccines cause disease?<br />

Some studies have suggested a temporal link between vaccinations and<br />

certain medical conditions, such as asthma, multiple sclerosis and diabetes.<br />

<strong>The</strong> questions of a link are often made for a disease of unknown cause. <strong>The</strong><br />

appearance of a certain medical condition after vaccination does not necessarily<br />

imply that they are causally related. Importantly, however, once an issue is raised<br />

it needs prompt research, discussion and then education to avoid creating a<br />

myth. In many cases, subsequent epidemiological studies have indicated that the<br />

association is due to chance alone. <strong>The</strong> following is a list of concerns that have<br />

been raised.<br />

482 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Does MMR vaccine cause inflammatory bowel disease or autistic spectrum disorder?<br />

No.<br />

In 1998, Wakefield et al. (Royal Free Hospital, London) published a case-series<br />

study with 12 children suggesting that MMR vaccine caused inflammatory bowel<br />

disease (IBD), which then resulted in decreased absorption of essential vitamins<br />

and nutrients through the intestinal tract. <strong>The</strong>y proposed that this could result in<br />

developmental disorders such as autism. <strong>The</strong> Lancet retracted this publication in<br />

2010 and the British Medical Council struck off the lead author in 2010, following<br />

the British General Medical Council’s Fitness to Practice Panel finding the author<br />

‘guilty of serious professional misconduct’.<br />

An extensive review published in 2004 by the Institute of Medicine (IOM),<br />

an independent expert body in the United States, concluded that there is no<br />

association between the MMR vaccine and the development of autism. A 2011<br />

update by the IOM continues to reject any causal association between MMR<br />

vaccine and autism (see www.iom.edu/vaccineadverseeffects).<br />

See 4.9 Measles for further information. <strong>The</strong>re is also an MMR vaccine decision<br />

aid designed for parents available at www.ncirs.edu.au/decisionaid/index.html.<br />

Do childhood immunisations cause asthma?<br />

<strong>The</strong>re is no evidence that vaccination causes or worsens asthma. It is especially<br />

important that children with asthma be vaccinated like other children, as<br />

catching a disease like whooping cough can make an asthma attack worse.<br />

Although influenza vaccine is not routinely recommended for all asthmatics,<br />

it is recommended for severe asthmatics, such as those requiring frequent<br />

hospitalisation (see 4.7 Influenza).<br />

Does influenza vaccine cause flu?<br />

No. It is not possible for influenza vaccine to cause ‘flu’ as it is not a live viral<br />

vaccine. (Note: a live attenuated influenza vaccine is used in some countries, but<br />

not in Australia.) As some people experience side effects such as a mild fever<br />

after the vaccine, it is understandable that they may confuse these symptoms<br />

with actually having the flu. In addition, the influenza vaccine is recommended<br />

to be given at the commencement of the flu season. Hence, it is possible that<br />

a person who has contracted, and is incubating, influenza during vaccination<br />

will mistakenly believe the vaccine to be causal. In addition, influenza vaccine<br />

is given at the very time of year when there are a lot of upper respiratory tract<br />

infections (URTIs) around. It is not uncommon for someone to attribute an URTI<br />

within a week of an influenza vaccine to the vaccine dose. Importantly, URTI<br />

symptoms occurring after influenza vaccine should not put people off having the<br />

vaccine the following year.<br />

APPENDIX 4 483<br />

APPENDIX 4


A4.4 Questions about vaccine content 4<br />

See also Table A3.1 Components of vaccines used in the National <strong>Immunisation</strong><br />

Program in Appendix 3. Refer also to the product information (PI) or the<br />

consumer medicines information (CMI) for individual vaccines; both are<br />

available from the TGA website (www.tga.gov.au).<br />

Preservatives<br />

Preservatives are used to prevent fungal and or bacterial contamination of the<br />

vaccine. <strong>The</strong>y include thiomersal, phenoxyethanol and phenol.<br />

Thiomersal<br />

Thiomersal (or thimerosal) is a compound that is partly composed of a form of<br />

mercury called ethylmercury. It has been used in very small amounts in vaccines<br />

for about 60 years to prevent bacterial and fungal contamination of vaccines. In<br />

the past, the small amount of thiomersal in vaccines was one of several potential<br />

sources of mercury. Diet (such as some seafood) and other environmental sources<br />

are also possible sources of mercury. Vaccines used in the past, such as DTP,<br />

contained only 25 µg of thiomersal per dose.<br />

Mercury causes a toxic effect after it reaches a certain level in the body. Whether<br />

or not it reaches a toxic level depends on the amount of mercury consumed and<br />

the person’s body weight; individuals with very low body weight are usually<br />

more susceptible to toxic effects from a certain intake of mercury. Thus, the<br />

possibility existed that vaccination of newborn babies, particularly those of very<br />

low birth weight, with repeated doses of thiomersal-containing vaccines might<br />

have resulted in levels of mercury above the recommended guidelines.<br />

Thiomersal was removed from vaccines in response to the above theoretical<br />

concern and to reduce total exposure to mercury in babies and young children in<br />

a world where other environmental sources may be more difficult to eliminate. 5-7<br />

Currently, all vaccines on the NIP for children and adolescents are free of<br />

thiomersal.<br />

Phenoxyethanol<br />

<strong>The</strong> aromatic ether alcohol, 2-phenoxyethanol, is used as a preservative in many<br />

vaccines, and also as a preservative in cosmetics. It is used in vaccines as an<br />

alternative preservative to thiomersal.<br />

Phenol<br />

Phenol is an aromatic alcohol used as a preservative in a few vaccines.<br />

Adjuvants<br />

Adjuvants are compounds used to enhance the immune response to vaccination<br />

and include various aluminium salts, such as aluminium hydroxide, aluminium<br />

phosphate and potassium aluminium sulphate (alum). A review of all available<br />

studies of aluminium-containing diphtheria, tetanus and pertussis vaccines<br />

484 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


(either alone or in combination) found no evidence that aluminium salts in<br />

vaccines cause any serious or long-term adverse events. 8<br />

Aluminium<br />

A small amount of aluminium salts has been added to some vaccines for<br />

about 60 years. Aluminium acts as an adjuvant, which improves the protective<br />

response to vaccination by keeping antigens near the injection site so they can<br />

be readily accessed by cells responsible for inducing an immune response. <strong>The</strong><br />

use of aluminium in vaccines means that, for a given immune response, less<br />

antigen is needed per dose of vaccine, and a lower number of total doses are<br />

required. Although aluminium-containing vaccines have been associated with<br />

local reactions and, less often, with the development of subcutaneous nodules at<br />

the injection site, other studies have reported fewer reactions with aluminiumadsorbed<br />

vaccines than with unadsorbed vaccines. Concerns about the longerterm<br />

effects of aluminium in vaccines arose after some studies suggested a link<br />

between aluminium in the water supply and Alzheimer’s disease, but this link<br />

has never been substantiated. <strong>The</strong> amount of aluminium in vaccines is very<br />

small and the intake from vaccines is far less than that received from diet or<br />

medications such as some antacids. 9,10<br />

Additives<br />

Additives are used to stabilise vaccines in adverse conditions (temperature<br />

extremes of heat and freeze drying) and to prevent the vaccine components<br />

adhering to the side of the vial.<br />

Examples of additives include:<br />

• lactose and sucrose (both sugars)<br />

• sorbitol and mannitol (both sugar alcohols)<br />

• polysorbate 80, made from sorbitol and oleic acid (an omega fatty acid)<br />

• glycine and monosodium glutamate or MSG (both are amino acids or salts of<br />

amino acids)<br />

• gelatin, which is partially hydrolysed collagen, usually of bovine or porcine<br />

origin, although information on the source of gelatin is not routinely<br />

provided in the product information for all vaccines.<br />

Some members of the Islamic and Jewish faiths may object to vaccination,<br />

arguing that vaccines can contain pork products. However, scholars of<br />

the Islamic Organization for Medical Sciences have determined that the<br />

transformation of pork products into gelatin will sufficiently alter them,<br />

thus making it permissible for observant Muslims to receive vaccines, even<br />

if the vaccines contain porcine gelatin. Likewise, leaders of the Jewish faith<br />

have also indicated that pork-derived additives to medicines are permitted.<br />

Further information may be obtained from the following websites<br />

APPENDIX 4 485<br />

APPENDIX 4


www.vaccinesafety.edu/Porcine-vaccineapproval.htm and www.immunize.<br />

org/concerns/porcine.pdf<br />

• human serum albumin (protein).<br />

Manufacturing residuals<br />

Manufacturing residuals are residual quantities of reagents used in the<br />

manufacturing process of individual vaccines. <strong>The</strong>y include antibiotics (such<br />

as neomycin or polymyxin), inactivating agents (e.g. formaldehyde) as well<br />

as cellular residuals (egg and yeast proteins), traces of which may be present<br />

in the final vaccine. Antibiotics are used during the manufacturing process to<br />

ensure that bacterial contamination does not occur; traces of these antibiotics<br />

may remain in the final vaccine. Inactivating agents are used to ensure that<br />

the bacterial toxin or viral components of the vaccine are not harmful, but will<br />

result in an immune response. Cellular residuals are minimised by extensive<br />

filtering. However, trace amounts may be present in the final product. <strong>The</strong> most<br />

commonly found residual is formaldehyde.<br />

Formaldehyde<br />

Formaldehyde is used during the manufacture of many vaccines. For example,<br />

with tetanus vaccines, formaldehyde is used to detoxify the tetanus toxin protein<br />

produced. <strong>The</strong> non-toxic protein, which becomes the active ingredient of the<br />

vaccine, is further purified to remove contaminants and any excess (unreacted or<br />

unbound) formaldehyde. <strong>The</strong> current standard applicable to vaccines for human<br />

use in Australia is less than 0.02% w/v of free formaldehyde. <strong>The</strong> maximum<br />

amount of free formaldehyde detected by the <strong>The</strong>rapeutic Goods Administration<br />

during testing of vaccines registered in Australia has been 0.004% w/v, which is<br />

well below the standard limit.<br />

Other ingredients and information about manufacturing<br />

Vaccines also may be made up in sterile water or sterile saline (salt-water).<br />

Some viruses used in vaccines require the use of ‘cell lines’ in which to grow the<br />

vaccine virus. <strong>The</strong> cell lines are not included as a component of the vaccine. Some<br />

of these cell lines (called human diploid cell lines – WI-38 and MRC-5) were<br />

originally derived from human fetal tissue in the 1960s. <strong>The</strong>se cell lines have<br />

been growing under laboratory conditions for more than 40 years, and there has<br />

been no further fetal tissue obtained since the 1960s. <strong>The</strong> vaccines manufactured<br />

using viruses that were grown in these cell lines include rubella vaccine and<br />

MMR vaccine, hepatitis A vaccines, varicella vaccines, rabies vaccine and oral<br />

polio (Sabin) vaccine (no longer available in Australia). Many of these vaccines<br />

prevent severe disease in unborn babies and infants, including, most notably,<br />

rubella, which causes congenital rubella syndrome.<br />

486 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


A4.5 Questions about the need for immunisation<br />

Isn’t natural immunity better than immunity from vaccination?<br />

While vaccine-induced immunity may diminish with time without boosters<br />

(vaccine or contact with wild-type infection), ‘natural’ immunity, acquired by<br />

catching the disease, is usually life-long, with the exception of pertussis. <strong>The</strong><br />

problem is that the wild or ‘natural’ disease has a higher risk of serious illness<br />

and occasionally death. Children or adults can be revaccinated (with some, but<br />

not all, vaccines) if their immunity from the vaccines falls to a low level or if<br />

previous research has shown that a booster vaccination is required for long-term<br />

protection. It is important to remember that vaccines are many times safer than<br />

the diseases they prevent.<br />

Diseases like measles, polio and diphtheria have already disappeared from<br />

most parts of Australia. Why do we need to keep vaccinating children against<br />

these diseases?<br />

Although these diseases are much less common now, they still exist. <strong>The</strong><br />

potential problem of disease escalation is kept in check by routine vaccination<br />

programs. In countries where vaccination rates have declined, vaccinepreventable<br />

diseases have sometimes reappeared. For example, Holland has one<br />

of the highest rates of fully vaccinated people in the world. However, in the early<br />

1990s, there was a large outbreak of polio among a group of Dutch people who<br />

belonged to a religious group that objected to vaccination. While many of these<br />

people suffered severe complications like paralysis, polio did not spread into the<br />

rest of the Dutch community. This was due to the high rate of vaccination against<br />

polio, which protected the rest of the Dutch community.<br />

<strong>The</strong>re have been recent outbreaks of whooping cough, measles and rubella<br />

in Australia, and a number of children have died. Cases of tetanus and<br />

diphtheria, although rare, still occur. Thus, even though these diseases are<br />

much less common now than in the past, it is necessary to continue to protect<br />

<strong>Australian</strong> children, so that the diseases cannot re-emerge to cause large<br />

epidemics and deaths.<br />

Also, many of the diseases against which we vaccinate our children are still<br />

common in other areas of the world. For example, measles still occurs in many<br />

Asian countries, where many people take holidays or travel for business.<br />

<strong>The</strong>refore, it is possible for non-immune individuals to acquire measles<br />

overseas, and, with the speed of air travel, arrive home and be able to pass<br />

measles onto those around them if they are unprotected. Measles is highly<br />

infectious and can infect others for several hours after an infected person has<br />

left a room. Vaccination, while not 100% effective, can considerably minimise<br />

a person’s chance of catching a disease. <strong>The</strong> more people who are vaccinated,<br />

the less chance there is that a disease, such as measles, will spread widely in the<br />

community. This is referred to as ‘herd immunity’.<br />

APPENDIX 4 487<br />

APPENDIX 4


Why do some children get the disease despite being vaccinated?<br />

This is possible because a small proportion of those who are vaccinated will<br />

remain susceptible to the disease. However, in the cases in which illness does<br />

occur in vaccinated individuals, the illness is usually much less severe than in<br />

those who were not vaccinated. <strong>The</strong> protection provided by the same vaccine<br />

to different individuals can differ. For example, if 100 children are vaccinated<br />

with MMR, 5 to 10 of the 100 fully vaccinated children might still catch measles,<br />

mumps or rubella (although the disease will often be less severe in vaccinated<br />

children). If 100 children are vaccinated with a full schedule of pertussiscontaining<br />

vaccines, 20 of the children might still get whooping cough, but,<br />

once again, the disease is often less severe in these vaccinated children. To put<br />

it another way, if you do not vaccinate 100 children with MMR vaccine, and the<br />

children are exposed to measles, all of them will catch the disease with a risk<br />

of high rates of complications like pneumonia or encephalitis. <strong>The</strong> reason why<br />

fewer children become infected than these figures suggest is due to the high<br />

vaccine coverage rates in the community. If there are high coverage rates, there<br />

is less chance of contact with the infection and, although some children may be<br />

susceptible, they have a low chance of contact with the infection (this situation is<br />

also called ‘herd immunity’).<br />

What about homeopathic ‘immunisation’?<br />

Homeopathic ‘immunisation’ has not been proved to give protection against<br />

infectious diseases; only conventional vaccination produces a measurable<br />

immune response. <strong>The</strong> Council of the Faculty of Homeopathy, London, issued a<br />

statement in 1993, which reads: ‘<strong>The</strong> Faculty of Homeopathy, London, strongly<br />

supports the conventional vaccination program and has stated that vaccination<br />

should be carried out in the normal way, using the conventional tested and<br />

proved vaccines, in the absence of medical contraindications’. 11<br />

A4.6 Further information about vaccination<br />

More information about vaccination can be found in the following publications<br />

produced by the <strong>Australian</strong> Government Department of Health and Ageing:<br />

• Understanding childhood immunisation<br />

• <strong>Immunisation</strong> myths and realities – responding to arguments against immunisation:<br />

a guide for providers.<br />

<strong>The</strong> following two websites include further publications, fact sheets, etc. and are<br />

recommended for both immunisation service providers and the general public:<br />

• Immunise Australia website www.immunise.health.gov.au<br />

• <strong>The</strong> National Centre for <strong>Immunisation</strong> Research and Surveillance of Vaccine<br />

Preventable Diseases (NCIRS) website www.ncirs.edu.au.<br />

Also, check with your local state or territory Public Health Unit or local<br />

council, maternal child health nurse or public health vaccination clinic for more<br />

information (see Appendix 1 Contact details for <strong>Australian</strong>, state and territory<br />

government health authorities and communicable disease control).<br />

References<br />

A full reference list is available on the electronic <strong>Handbook</strong> or website<br />

www.immunise.health.gov.au<br />

488 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


APPENDIX 5: GLOSSARY OF TECHNICAL TERMS<br />

Adjuvant<br />

a preparation added to a vaccine to improve the immune response to that vaccine<br />

Adverse event following immunisation (AEFI)<br />

an unwanted reaction following administration of a vaccine, which may or may<br />

not be caused by the vaccine; adverse events may be at the site of injection, or<br />

may be a general illness or a general allergic reaction<br />

Anaphylaxis<br />

a sudden and severe allergic reaction, which results in a serious fall in blood<br />

pressure and/or respiratory obstruction and may cause unconsciousness and<br />

death if not treated immediately<br />

Attenuation<br />

the process of modifying a virus or bacteria to reduce its virulence (diseaseinducing<br />

ability) while retaining its ability to induce a strong immune response<br />

(immunogenicity)<br />

Bacteria<br />

microorganisms that are smaller than a blood cell, but bigger than a virus;<br />

examples of bacterial infections are diphtheria, tetanus, pertussis, Hib and<br />

tuberculosis<br />

Brachial neuritis<br />

pain in the arm, causing persisting weakness of the limb on the side of<br />

vaccination<br />

Chronically infected<br />

formerly referred to as a ‘carrier’; a person who has an infection that, although<br />

not necessarily causing symptoms, may still be active and may spread to others;<br />

chronic infection may last for years; examples of infections that can result in<br />

chronically infected states are hepatitis B and typhoid<br />

Conjugate<br />

some bacterial vaccines (e.g. Hib, meningococcal and pneumococcal conjugate<br />

vaccines) are made from the chemical linking (conjugation) of a tiny amount of<br />

the ‘sugar’ (correctly known as the polysaccharide) that makes up the cell coat of<br />

the bacteria with a protein molecule, in order to improve the immune response to<br />

the vaccine<br />

Contraindication<br />

a reason why a vaccine or drug must not be given<br />

Corticosteroid<br />

a drug used to reduce inflammation and other immune responses<br />

APPENDIX 5 489<br />

APPENDIX 5


DT<br />

a vaccine that protects against diphtheria and tetanus. <strong>The</strong> acronym DT,<br />

using capital letters, signifies the child formulation of diphtheria and tetanuscontaining<br />

vaccine, and denotes the substantially larger amounts of diphtheria<br />

toxoid in this formulation than in the adolescent/adult formulation.<br />

dT<br />

reduced antigen content formulation of diphtheria-tetanus vaccine, which<br />

contains substantially lower concentrations of diphtheria toxoid, and<br />

approximately half the tetanus antigen content, than the child formulation<br />

(which is signified by using capital letters DT). This vaccine is most commonly<br />

administered to adolescents/adults.<br />

DTP/DTPa/DTPw<br />

a vaccine that protects against diphtheria, tetanus and pertussis (whooping<br />

cough). <strong>The</strong> DTP used in Australia and many other industrialised countries is<br />

DTPa, which contains an acellular pertussis component made of refined pertussis<br />

extracts instead of inactivated whole pertussis bacteria (DTPw). <strong>The</strong> acronym<br />

DTPa, using capital letters, signifies child formulations of diphtheria, tetanus<br />

and acellular pertussis-containing vaccines, and denotes the substantially larger<br />

amounts of diphtheria toxoid and pertussis antigens in these formulations than<br />

in the adolescent/adult formulations.<br />

dTpa<br />

reduced antigen content formulation of diphtheria-tetanus-acellular pertussis<br />

vaccine, which contains substantially lower concentrations of diphtheria toxoid<br />

and pertussis antigens, and approximately half the tetanus antigen content, than<br />

the child formulations (which are signified by using all capital letters [DTPa]).<br />

This vaccine is most commonly administered to adolescents/adults.<br />

Effectiveness<br />

the extent to which a vaccine produces a benefit in a defined population in<br />

uncontrolled or routine circumstances<br />

Efficacy<br />

the extent to which a vaccine produces a benefit in a defined population in<br />

controlled or ideal circumstances, for example, in a randomised controlled trial<br />

Encephalitis<br />

inflammation of the brain<br />

Encephalopathy<br />

a general term to describe a variety of illnesses that affect the brain, including<br />

encephalitis<br />

Endemic<br />

endemic infections are present all the time in a community<br />

490 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Enzootic<br />

enzootic infections are present all the time in animals of a specific geographic<br />

area<br />

Epidemic<br />

epidemic infections are those that spread rapidly in a community; measles and<br />

influenza viruses are common causes of epidemics in Australia; small epidemics<br />

are often called outbreaks<br />

Extensive limb swelling<br />

swelling of the limb, with or without redness, which:<br />

» extends from the joint above to the joint below the injection site, or beyond<br />

a joint (above or below the injection site), or<br />

» results in the circumference of the limb being twice the normal size.<br />

Febrile<br />

related to a fever, as in febrile illness and febrile convulsions<br />

Hepatitis<br />

an inflammation of the liver; can be caused by viral infections<br />

Hypotonic-hyporesponsive episode (shock, collapse)<br />

the sudden onset of pallor or cyanosis, limpness (muscle hypotonia), and<br />

reduced responsiveness or unresponsiveness occurring after vaccination,<br />

where no other cause is evident, such as a vasovagal episode or anaphylaxis.<br />

<strong>The</strong> episode usually occurs 1 to 48 hours after vaccination and resolves<br />

spontaneously.<br />

<strong>Immunisation</strong><br />

the process of inducing immunity to an infectious agent by administering a<br />

vaccine<br />

Immunity<br />

the ability of the body to fight off certain infections; immunity can result from<br />

natural (‘wild’) infections or from vaccination<br />

Immunogenicity<br />

the ability (or the degree) to which a particular substance, in this context a<br />

vaccine, may provoke an immune response<br />

Immunoglobulin<br />

a protein extract from blood, sometimes called ‘antibody’, that fights off infection;<br />

injection of immunoglobulins provides temporary immunity against certain<br />

infections<br />

APPENDIX 5 491<br />

APPENDIX 5


Incubation period<br />

after a person is infected with bacteria or viruses, it often takes days or weeks<br />

for the infection to cause an obvious illness; the time between exposure to the<br />

infectious agent and development of the disease is called the incubation period<br />

Infection<br />

an infection occurs when bacteria or viruses invade the body; if the body cannot<br />

fight the infection, it may cause an illness<br />

Intradermal (ID) injection<br />

an injection into the surface layers of the skin; this is used for the administration<br />

of bacille Calmette-Guérin (BCG), the tuberculosis vaccine<br />

Intramuscular (IM) injection<br />

an injection into the muscle; vaccines are usually injected into a muscle of the<br />

upper outer thigh, or a muscle in the upper arm<br />

Intussusception<br />

when one portion of the bowel telescopes into the next portion of bowel,<br />

resulting in a blockage<br />

Invasive disease<br />

this term is often used when talking about pneumococcal or meningococcal<br />

disease. This term means that the bacteria (or germs) have been found in the<br />

blood, spinal fluid or another part of the body that would normally be sterile (or<br />

germ free).<br />

Jaundice<br />

yellow skin colour that may result from severe hepatitis<br />

Pandemic influenza<br />

a global epidemic that results when a new strain of influenza virus appears in the<br />

human population. It causes more severe disease in the population because there<br />

is little immunity to this new strain.<br />

Paracetamol<br />

a medicine that helps reduce fever; it may be given to minimise fevers following<br />

vaccination<br />

Pertussis<br />

whooping cough, an illness caused by a bacterium, Bordetella pertussis<br />

Polysaccharide<br />

a group of complex carbohydrates (sugars), which make up the cell coating<br />

present in some bacteria<br />

Polyvalent vaccine<br />

a combination vaccine that protects against more than one disease; examples are<br />

DTPa and MMR<br />

492 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Rotavirus<br />

a virus that is a common cause of diarrhoea (and often vomiting as well) in<br />

young children. <strong>The</strong> diarrhoea can be severe in very young children, such that<br />

they may need intravenous fluids (i.e. through a vein in the arm) in hospital.<br />

Rubella<br />

a viral illness, sometimes also known as German measles<br />

Seizure<br />

a witnessed sudden loss of consciousness and generalised, tonic, clonic, tonic–<br />

clonic, or atonic motor manifestations.<br />

Types of seizures include:<br />

» febrile seizures; with fever >38.5°C<br />

» afebrile seizures; without fever<br />

» syncopal seizures; a syncope/vasovagal episode followed by seizure(s).<br />

Subcutaneous (SC) injection<br />

an injection into the tissue between the skin and the underlying muscle<br />

Syncope<br />

see vasovagal episode<br />

Thrombocytopenia<br />

platelet count


Virus<br />

a tiny living organism, smaller than a bacterium, that can cause infections;<br />

measles, rubella, mumps, polio, influenza and hepatitis B are examples of viruses<br />

Zoster<br />

an abbreviation for herpes zoster infection (also known as shingles); a painful<br />

rash and illness, caused by the varicella-zoster (chickenpox) virus<br />

494 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


APPENDIX 6: COMMONLY USED ABBREVIATIONS<br />

ABLV <strong>Australian</strong> bat lyssavirus<br />

ACIR <strong>Australian</strong> Childhood <strong>Immunisation</strong> Register<br />

ACT <strong>Australian</strong> Capital Territory<br />

ADRS Adverse Drug Reactions System<br />

AEFI adverse event following immunisation<br />

AIDS acquired immunodeficiency syndrome<br />

anti-HBe antibody to hepatitis B e antigen<br />

anti-HBc antibody to hepatitis B core antigen<br />

anti-HBs antibody to hepatitis B surface antigen<br />

AOM acute otitis media<br />

ASCIA Australasian Society of Clinical Immunology and Allergy<br />

ATAGI <strong>Australian</strong> Technical Advisory Group on <strong>Immunisation</strong><br />

BCG bacille Calmette-Guérin<br />

CCID50 cell culture infectious dose 50%<br />

CDNA Communicable Diseases Network Australia<br />

CI confidence interval<br />

CIN cervical intraepithelial neoplasia<br />

CRS congenital rubella syndrome<br />

CSF cerebrospinal fluid<br />

DNA deoxyribonucleic acid<br />

DT diphtheria-tetanus vaccine for use in children (for further<br />

explanation of this term, see Appendix 5 Glossary of technical terms)<br />

dT diphtheria-tetanus vaccine for use in adults (for further<br />

explanation of this term, see Appendix 5 Glossary of technical terms)<br />

DTPa diphtheria-tetanus-acellular pertussis vaccine (for further<br />

explanation of this term, see Appendix 5 Glossary of technical terms)<br />

dTpa diphtheria-tetanus-acellular pertussis vaccine, reduced antigen<br />

content formulation (for further explanation of this term, see<br />

Appendix 5 Glossary of technical terms)<br />

DTPw diphtheria-tetanus-whole-cell pertussis vaccine (for further<br />

explanation of this term, see Appendix 5 Glossary of technical terms)<br />

EIA enzyme immunoassay<br />

ELISA enzyme-linked immunosorbent assay<br />

FHA filamentous haemagglutinin<br />

FIM fimbriae (pertussis)<br />

GBS Guillain-Barré syndrome<br />

GP general practitioner<br />

GVHD graft-versus-host disease<br />

HAV hepatitis A virus<br />

HBcAg hepatitis B core antigen<br />

HBeAg hepatitis B e antigen<br />

APPENDIX 6 495<br />

APPENDIX 6


HBIG hepatitis B immunoglobulin<br />

HBsAg hepatitis B surface antigen<br />

HBV hepatitis B virus<br />

HCW healthcare worker<br />

HDCV human diploid cell vaccine (rabies)<br />

HepA hepatitis A vaccine<br />

HepB hepatitis B vaccine<br />

HHE hypotonic-hyporesponsive episode<br />

Hib Haemophilus influenzae type b<br />

Hib-MenCCV Haemophilus influenzae type b-meningococcal C conjugate vaccine<br />

HIV human immunodeficiency virus<br />

HPV human papillomavirus<br />

2vHPV bivalent HPV vaccine<br />

4vHPV quadrivalent HPV vaccine<br />

HRIG human rabies immunoglobulin<br />

HSCT haematopoietic stem cell transplant<br />

HZ herpes zoster<br />

ID intradermal<br />

IgA/G/M immunoglobulin A/G/M<br />

IM intramuscular<br />

IPD invasive pneumococcal disease<br />

IPV inactivated poliomyelitis vaccine<br />

IS intussusception<br />

ITP idiopathic thrombocytopenia purpura<br />

IU international units<br />

IV intravenous<br />

JE Japanese encephalitis<br />

LT-ETEC heat-labile toxin producing enterotoxigenic Escherichia coli<br />

MenCCV meningococcal serogroup C conjugate vaccine<br />

4vMenCV quadrivalent meningococcal conjugate vaccine<br />

4vMenPV quadrivalent meningococcal polysaccharide vaccine<br />

MMR measles-mumps-rubella<br />

MMRV measles-mumps-rubella-varicella<br />

NCIRS National Centre for <strong>Immunisation</strong> Research and Surveillance of<br />

Vaccine Preventable Diseases<br />

NHIG normal human immunoglobulin<br />

NHMRC National Health and Medical Research Council<br />

NHVPR National HPV Vaccination Program Register<br />

NIP National <strong>Immunisation</strong> Program<br />

NSW New South Wales<br />

NT Northern Territory<br />

NTHi non-typeable Haemophilus influenzae<br />

OMP outer membrane protein<br />

496 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


OPV oral poliomyelitis vaccine<br />

PCECV purified chick embryo cell vaccine (rabies)<br />

PCR polymerase chain reaction<br />

7vPCV 7-valent pneumococcal conjugate vaccine<br />

10vPCV 10-valent pneumococcal conjugate vaccine<br />

13vPCV 13-valent pneumococcal conjugate vaccine<br />

PEP post-exposure prophylaxis<br />

pH1N1 pandemic influenza A(H1N1)pdm09<br />

PHN post-herpetic neuralgia<br />

PI product information<br />

PreP pre-exposure prophylaxis<br />

23vPPV 23-valent pneumococcal polysaccharide vaccine<br />

PRN pertactin<br />

PRP polyribosylribitol phosphate<br />

PRP-OMP PRP conjugated to the outer membrane protein of Neisseria<br />

meningitidis<br />

PRP-T PRP conjugated to tetanus toxoid<br />

PT pertussis toxoid<br />

Qld Queensland<br />

RCT randomised controlled trial<br />

RIG rabies immunoglobulin<br />

RNA ribonucleic acid<br />

SA South Australia<br />

SC subcutaneous<br />

SCID severe combined immunodeficiency<br />

SIDS sudden infant death syndrome<br />

SOT solid organ transplant<br />

SSPE subacute sclerosing panencephalitis<br />

Tas Tasmania<br />

TB tuberculosis<br />

TCID50 tissue culture infectious dose 50%<br />

TGA <strong>The</strong>rapeutic Goods Administration<br />

TIG tetanus immunoglobulin<br />

TST tuberculin skin test<br />

Vic Victoria<br />

VLP virus-like particle<br />

VNAb (rabies) virus neutralising antibody<br />

VPD vaccine-preventable disease<br />

VV varicella vaccine<br />

VZV varicella-zoster virus<br />

WA Western Australia<br />

WHO World Health Organization<br />

ZIG zoster immunoglobulin<br />

APPENDIX 6 497<br />

APPENDIX 6


APPENDIX 7: OVERVIEW OF VACCINE AVAILABILITY<br />

IN AUSTRALIA<br />

Table A7.1 provides an overview of key dates when vaccines first came into<br />

widespread use in Australia. This table provides only some dates; for specific<br />

details on vaccine registration, funding, recommendations and program use,<br />

please see complete information in the ‘NCIRS Vaccination History Tables’<br />

available from the NCIRS website (www.ncirs.edu.au/immunisation/history/<br />

index.php). <strong>The</strong>se tables, listed by vaccine type, provide a summary of the<br />

significant events in vaccination practice in Australia, particularly for vaccines<br />

used in population-based immunisation programs. Additional information<br />

regarding vaccines available in each jurisdiction can also be sought from your<br />

local state or territory <strong>Immunisation</strong> Department.<br />

Table A7.1: Key dates when vaccines first came into widespread use in Australia<br />

Year Vaccine<br />

1945 Tetanus toxoid<br />

1953 Diphtheria-tetanus-pertussis, whole-cell (DTPw)<br />

1956 Poliomyelitis (Salk) (inactivated poliomyelitis vaccine [IPV])<br />

1966 Poliomyelitis (Sabin) (live attenuated oral poliomyelitis vaccine [OPV])<br />

1970 Measles<br />

1971 Rubella<br />

1975 Child diphtheria-tetanus (CDT)<br />

1982 Adult diphtheria-tetanus (ADT)<br />

1982 Measles-mumps<br />

1982 Hepatitis B (hepB) (serum-derived vaccine)<br />

1987 Hepatitis B (recombinant vaccine)<br />

1989 Measles-mumps-rubella (MMR)<br />

1993 Hib (Haemophilus influenzae type b)<br />

1994 Hepatitis A<br />

1997 Diphtheria-tetanus-pertussis, acellular (DTPa)<br />

1999 Influenza<br />

1999 23-valent pneumococcal polysaccharide (23vPPV)<br />

2000 DTPa-hepB<br />

2000 Hib(PRP-OMP)-hepB<br />

2001 7-valent pneumococcal conjugate (7vPCV)<br />

498 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


Year Vaccine<br />

2003 Varicella<br />

2003 Meningococcal C conjugate<br />

2004 Diphtheria-tetanus-pertussis, acellular; reduced antigen content<br />

formulations (dTpa and dTpa-IPV)<br />

2005 Pentavalent and hexavalent combination DTPa vaccines (DTPa-hepB-IPV-<br />

Hib; DTPa-IPV; DTPa-hepB-IPV; DTPa-IPV-Hib)<br />

2007 Human papillomavirus (HPV)<br />

2007 Rotavirus<br />

2009 10-valent pneumococcal conjugate (10vPCV)<br />

2011 13-valent pneumococcal conjugate (13vPCV)<br />

<strong>2013</strong> Measles-mumps-rubella-varicella (MMRV)<br />

APPENDIX 7 499<br />

APPENDIX 7


INDEX<br />

Italicised numbers indicate tables<br />

2-phenoxyethanol, in vaccines, 19, 484<br />

7vPCV (7-valent pneumococcal conjugate<br />

vaccine), 45, 56–60, 108, 194, 320, 321, 322,<br />

328, 333<br />

adverse events following immunisation<br />

(AEFI), 335<br />

pneumococcal disease epidemiology,<br />

318–319<br />

10vPCV (10-valent pneumococcal conjugate<br />

vaccine), 8, 14, 43, 46, 52–53, 56, 58, 60, 68,<br />

194, 319, 320. see also pneumococcal disease<br />

adverse events following immunisation<br />

(AEFI), 335<br />

dosage and administration, 323–324<br />

and Indigenous children, 108<br />

pregnancy and breastfeeding, 334<br />

route of vaccine administration, 68–69<br />

transport, storage and handling, 323<br />

vaccination recommendations, 324–333<br />

13vPCV (13-valent pneumococcal conjugate<br />

vaccine), 8, 14–15, 43, 46, 62, 68, 105, 149, 151,<br />

156, 160, 160–161, 163–164, 174, 194, 251, 257,<br />

319–322<br />

adverse events following immunisation<br />

(AEFI), 334–335<br />

catch-up vaccination, 52–53, 57–58<br />

catch-up vaccination guidelines for<br />

children with medical condition<br />

associated with an increased risk, 59–60<br />

catch-up vaccination guidelines for<br />

children without medical condition<br />

associated with an increased risk, 56<br />

catch-up vaccination schedules for those<br />

≥10 years of age, 63–64<br />

dosage and administration, 323–324<br />

and Indigenous children, 108<br />

and influenza vaccination, 257<br />

minimum acceptable vaccine dose<br />

intervals, 50<br />

number of vaccine doses for children, 49<br />

precautions, 334<br />

pregnancy and breastfeeding, 136, 334<br />

vaccination recommendations, 324–333<br />

variations from product information,<br />

336–337<br />

23vPPV (23-valent pneumococcal<br />

polysaccharide vaccine), 15, 62, 105, 110, 137,<br />

143, 149, 151, 156, 160, 161, 163, 163–164, 321,<br />

321–322, 455, 471<br />

adverse events following immunisation<br />

(AEFI), 336<br />

catch-up vaccination guidelines for<br />

children with medical condition<br />

associated with an increased risk, 59–60<br />

500 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

catch-up vaccination schedules for those<br />

≥10 years of age, 63–64<br />

minimum acceptable vaccine dose<br />

intervals, 50<br />

number of vaccine doses for children, 49<br />

pneumococcal disease epidemiology,<br />

318–319<br />

route of vaccine administration, 68–69<br />

vaccination recommendations, 324–333<br />

A<br />

abattoir workers. see animals, persons working<br />

with<br />

abbreviations list, 495–497<br />

Aboriginal and Torres Strait Islander people,<br />

35, 104–112<br />

additional recommended vaccines, 105<br />

adults, 109–111<br />

catch-up vaccination, 57–58<br />

changes to recommendations in the <strong>10th</strong><br />

edition of the handbook, 9, 12, 15<br />

children, 49, 50, 106–108<br />

Haemophilus influenzae type b (Hib),<br />

106–107, 192<br />

hepatitis A, 47, 107, 198–199, 201, 203, 206<br />

hepatitis B, 109, 209, 221<br />

human papillomavirus (HPV), 233<br />

influenza, 108, 109–110, 246, 254<br />

Japanese encephalitis, 111<br />

pneumococcal disease, 52–53, 108, 110–111,<br />

133, 318–319, 321, 324–325, 327–328, 329,<br />

330–332<br />

rotavirus, 373<br />

rubella, 111, 390<br />

service delivery to, 112<br />

tuberculosis, 106, 408, 412<br />

Act-HIB, 51, 55, 192<br />

transport, storage and handling, 194<br />

variations from product information, 197<br />

active immunisation, 18–20<br />

acute febrile illness, vaccination<br />

recommendations, 31<br />

acute otitis media (AOM). see otitis media<br />

acute systemic illness, vaccination<br />

recommendations, 31<br />

Adacel, 123, 185, 306, 400<br />

vaccine components, 470–471<br />

variations from product information, 189,<br />

315, 406–407<br />

Adacel Polio, 185, 189, 306, 315, 340, 344, 400<br />

variations from product information, 407<br />

additives, in vaccines, 485–486<br />

adjuvants, in vaccines, 19, 484–485<br />

administration of vaccines, 65–84<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 9<br />

equipment, 65–69


identifying the injection site, 79–82<br />

injection techniques, 71–73<br />

multiple vaccine injections, 83–84<br />

occupational health and safety issues, 65<br />

positioning, 75–78<br />

preparation, 70<br />

recommended injection sites, 73–74<br />

route, 67–69<br />

adolescents<br />

catch-up vaccination schedules, 61–62,<br />

63–64<br />

diphtheria, 186<br />

hepatitis B, 213, 214, 214–215, 220<br />

human papillomavirus (HPV), 238<br />

injection sites, 74<br />

measles, post-exposure prophylaxis, 281<br />

measles, vaccination, 273<br />

multiple vaccine injections, 83–84<br />

mumps, 299–300<br />

pertussis, 306<br />

positioning for vaccination, 78<br />

Q fever, 348–350<br />

rabies, 356–357<br />

rubella, 389–390<br />

tetanus, 400, 401<br />

varicella, 425, 428–429, 429<br />

adrenaline<br />

autoinjectors, 90–91<br />

doses, 90<br />

in management of anaphylaxis, 89–91<br />

ADT Booster, 123, 184, 400<br />

variations from product information, 189,<br />

406<br />

adults<br />

catch-up vaccination schedules, 61–64,<br />

63–64<br />

cholera, 179<br />

consent on behalf of an adult lacking<br />

capacity, 27<br />

diphtheria, 186–187<br />

hepatitis A, 202<br />

hepatitis B, 213–214, 214–215, 220–224<br />

human papillomavirus (HPV), 238–239<br />

Indigenous persons, 109–111<br />

influenza, 251<br />

injection sites, 74<br />

measles, post-exposure prophylaxis, 281<br />

measles, vaccination, 273<br />

multiple vaccine injections, 83–84<br />

mumps, 299–300<br />

pertussis, 306, 308–309<br />

pneumococcal disease, 326–327, 330–333<br />

poliomyelitis, 342<br />

positioning for vaccination, 78<br />

Q fever, 348–350<br />

rabies, 356–357<br />

rubella, 389–390<br />

tetanus, 400, 402, 403–404<br />

vaccination status and history, 40<br />

varicella vaccination, 425, 429<br />

and varicella vaccination, 434<br />

yellow fever vaccination, 444<br />

and zoster, 446–447, 447–448<br />

zoster vaccination, 450–453<br />

adverse events following immunisation (AEFI),<br />

9, 18, 21–23, 85–97, 481–482<br />

anaphylaxis and vasovagal episodes, 87–91<br />

cholera, 181<br />

common events, 91<br />

diphtheria, 187–188<br />

events without causal link, 94<br />

Haemophilus influenzae type b (Hib), 196<br />

hepatitis A, 207<br />

hepatitis B, 227–228<br />

human papillomavirus (HPV), 240, 241–242<br />

influenza, 249, 257–258<br />

Japanese encephalitis, 265–266<br />

measles, 278–280<br />

meningococcal disease, 292<br />

mumps, 301<br />

mumps vaccination, 301<br />

passive immunisation, 458, 464<br />

pertussis, 311, 312–313<br />

pneumococcal disease, 335–336<br />

poliomyelitis, 343<br />

poliomyelitis vaccination, 343<br />

Q fever, 351–352<br />

rabies and other lyssaviruses (including<br />

<strong>Australian</strong> bat lyssavirus), 370–371<br />

reporting of, 94–97<br />

rotavirus, 381–382<br />

rubella, 394<br />

state and territory contact information, 96<br />

tetanus, 405<br />

tuberculosis (TB), 414<br />

typhoid, 422<br />

uncommon or rare events, 92–93<br />

vaccination of persons who have had an<br />

AEFI, 130–133<br />

varicella, 433–435<br />

yellow fever, 443, 444–445<br />

zoster (herpes zoster), 448, 454<br />

Aedes aegypti, 439. see also yellow fever<br />

Africa<br />

cholera, 176<br />

hepatitis B, 118, 209, 221<br />

influenza, 245<br />

measles, 268, 269<br />

meningococcal disease, 120, 284<br />

migrants to Australia from, 174<br />

poliomyelitis, 338<br />

rabies and other lyssaviruses (including<br />

<strong>Australian</strong> bat lyssavirus), 354, 361<br />

rubella, 390<br />

INDEX 501<br />

INDEX


travel to, 116<br />

tuberculosis, 408<br />

typhoid, 416, 418<br />

yellow fever, 114, 439–440, 443<br />

agammaglobulinaemia, vaccination<br />

recommendations, 166<br />

aged care facility residents, and influenza<br />

vaccination, 255<br />

agricultural staff and students, vaccination<br />

recommendations, 172<br />

Agrippal, 247, 470–472<br />

vaccine components, 469–472<br />

AIDS, and zoster vaccination, 450–451<br />

albumin, in vaccines, 469<br />

alcoholism, 13<br />

and influenza vaccination, 254<br />

and pneumococcal disease, 327<br />

and tuberculosis (TB), 408<br />

allergies, 32, 131–133, 480–481. see also egg<br />

allergies<br />

aluminium hydroxide, in vaccines, 19, 469–470,<br />

484–485<br />

aluminium phosphate, in vaccines, 470,<br />

484–485<br />

Americas<br />

cholera, 176<br />

hepatitis B, 118, 209<br />

poliomyelitis, 338<br />

rabies and other lyssaviruses (including<br />

<strong>Australian</strong> bat lyssavirus), 354, 361<br />

rubella, 390<br />

tuberculosis, 409<br />

yellow fever, 114, 116, 439, 440<br />

anaesthesia, vaccination recommendations<br />

before or after, 168<br />

anal cancer, 233<br />

anaphylaxis, 37, 43, 87–91, 92–93, 131–132<br />

adrenaline use, 89–91<br />

and cholera vaccination, 180<br />

clinical features, 88<br />

contraindications to vaccination, 37<br />

and diphtheria vaccination, 187, 190<br />

and Haemophilus influenzae type b (Hib)<br />

vaccination, 196, 197<br />

and hepatitis A vaccination, 206<br />

and hepatitis B vaccination, 227, 230<br />

and human papillomavirus vaccination,<br />

241–242<br />

and immunoglobulin injection, 464<br />

an influenza vaccination, 256–257, 258<br />

and Japanese encephalitis vaccination, 265<br />

and measles vaccination, 275, 279<br />

and meningococcal vaccination, 292<br />

and pertussis vaccination, 312<br />

and pneumococcal vaccination, 334<br />

and poliomyelitis vaccination, 343<br />

response kit, 24<br />

502 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

and rotavirus vaccination, 379<br />

and rubella vaccination, 392<br />

and tetanus vaccination, 404, 405<br />

and tuberculosis vaccination, 414<br />

and typhoid vaccination, 421<br />

vaccination recommendations, 32<br />

and varicella vaccination, 431<br />

and yellow fever vaccination, 443, 444<br />

and zoster vaccination, 453<br />

animals, persons working with<br />

influenza vaccination, 255<br />

Q fever vaccination, 346, 348<br />

vaccination recommendations, 172<br />

anogenital warts, 233–234<br />

anterolateral thigh injection site, 79–80<br />

antibiotics, and typhoid vaccination, 421<br />

antiviral medication, and zoster vaccination,<br />

454<br />

armed forces<br />

hepatitis B, 223<br />

typhoid, 420<br />

vaccination recommendations, 171<br />

Asia<br />

cholera, 176<br />

diphtheria, 187<br />

hepatitis B, 118, 209, 221<br />

influenza, 245<br />

Japanese encephalitis, 113–114, 119,<br />

259–260, 263–264<br />

measles, 268, 269, 487<br />

meningococcal disease, 284<br />

poliomyelitis, 338<br />

rabies and other lyssaviruses (including<br />

<strong>Australian</strong> bat lyssavirus), 345, 361<br />

rubella, 390<br />

tick-borne encephalitis, 121<br />

tuberculosis, 408<br />

typhoid, 122, 416<br />

yellow fever, 439<br />

aspirin therapy<br />

and influenza vaccination, 254<br />

and measles vaccination, 278<br />

and varicella vaccination, 433, 438<br />

and zoster vaccination, 454<br />

asplenia, persons with, 10<br />

Haemophilus influenzae type b (Hib)<br />

vaccination, 195–196, 197<br />

meningococcal vaccination, 52, 136, 137,<br />

153, 283, 290, 291, 294<br />

and pneumococcal disease, 317, 326–327,<br />

332<br />

vaccination recommendations, 35, 62,<br />

161–164, 163–164<br />

<strong>Australian</strong> bat lyssavirus (ABLV), 353, 354. see<br />

also rabies and other lyssaviruses (including<br />

<strong>Australian</strong> bat lyssavirus)


<strong>Australian</strong> Capital Territory<br />

adverse events following immunisation<br />

(AEFI) notification, 95, 96<br />

age of consent, 26<br />

<strong>Australian</strong> Red Cross Blood Service, 457<br />

government health authority and<br />

communicable disease control contact<br />

details, 465–466<br />

pneumococcal vaccination, 52, 56<br />

<strong>Australian</strong> Childhood <strong>Immunisation</strong> Register<br />

(ACIR), 39, 89–99, 174, 268, 465<br />

<strong>Australian</strong> Government Department of Health<br />

and Ageing, 4, 22, 122, 129, 442, 465, 488<br />

<strong>Australian</strong> Government Department of<br />

Immigration and Citizenship (DIAC),<br />

information about refugee immunisation, 173<br />

<strong>Australian</strong> Influenza Vaccine Committee, 248<br />

<strong>Australian</strong> Q Fever Register, 102, 349<br />

<strong>Australian</strong> Red Cross Blood Service, 457<br />

<strong>Australian</strong> Technical Advisory Group on<br />

<strong>Immunisation</strong>, xi–xii, 1, 3<br />

autism, 94, 280, 483<br />

autoimmune conditions, vaccination<br />

recommendations, 165<br />

Avaxim, 199<br />

lower age limits for, 127<br />

recommended doses and schedules, 125,<br />

202<br />

vaccine components, 469–471<br />

B<br />

BabyBIG, 461<br />

bacille Calmette-Guérin vaccine. see BCG<br />

vaccine<br />

bats. see animals, persons working with<br />

BCG (bacille Calmette-Guérin vaccine) vaccine,<br />

16, 37, 67, 409–410<br />

adverse events following immunisation<br />

(AEFI), 414<br />

contraindications to vaccination, 413<br />

dosage and administration, 410–411<br />

and HIV-infected persons, 159<br />

and immunocompromised persons,<br />

147–148<br />

for Indigenous neonates, 104, 105, 106<br />

precautions, 414<br />

and pregnancy, 139, 142, 413<br />

recipients of, further vaccination<br />

recommendations, 32<br />

route of vaccine administration, 68–69, 72<br />

and subsequent live vaccines, 33, 271, 276,<br />

298, 300, 376, 388, 393<br />

transport, storage and handling, 410<br />

and travellers, 121<br />

vaccination procedures, 411<br />

vaccination recommendations, 412–413<br />

variations from product information, 414<br />

bleeding disorders, vaccination<br />

recommendations, 35, 168<br />

blood product recipients, 33, 166–167<br />

hepatitis B, 222–223<br />

measles, 276–277<br />

rotavirus, 381<br />

rubella, 393–394<br />

varicella, 432–433<br />

zoster, 454<br />

body-piercers. see skin penetration procedures<br />

bone marrow malignancies, and tuberculosis<br />

vaccination, 413<br />

Boostrix, 185, 306, 400<br />

doses and route of administration, 123<br />

vaccine components, 470<br />

variations from product information, 189,<br />

190, 315–316, 406–407<br />

Boostrix-IPV, 185, 307, 341, 400<br />

doses and route of administration, 123<br />

variations from product information, 189,<br />

190, 315–316, 344, 407<br />

borax, in vaccines, 470<br />

Bordetella parapertussis, 302. see also pertussis<br />

Bordetella pertussis, 302, 398. see also pertussis<br />

botulism antitoxin, 461<br />

availability, 457<br />

brachial neuritis, 93, 489<br />

and diphtheria vaccination, 187–188<br />

and pertussis vaccination, 313<br />

and tetanus vaccination, 405<br />

breast cancer patients, injection sites, 74<br />

breastfeeding<br />

and cholera vaccination, 180<br />

and diphtheria vaccination, 187<br />

and Haemophilus influenzae type b (Hib)<br />

vaccination, 196<br />

and hepatitis A vaccination, 206<br />

and human papillomavirus (HPV)<br />

vaccination, 240<br />

and influenza vaccination, 256<br />

and Japanese encephalitis vaccination,<br />

264–265<br />

and measles vaccination, 275<br />

and meningococcal vaccination, 291–292<br />

and mumps vaccination, 300<br />

and pertussis vaccination, 310–311<br />

and pneumococcal vaccination, 334<br />

and Q fever vaccination, 350–351<br />

and rabies vaccination, 370<br />

and rotavirus vaccination, 379<br />

and rubella vaccination, 391–392<br />

and tetanus vaccination, 402<br />

and tuberculosis vaccination, 413<br />

vaccination recommendations, 142–143<br />

and varicella vaccination, 430–431<br />

and yellow fever vaccination, 443<br />

and zoster vaccination, 453<br />

INDEX 503<br />

INDEX


C<br />

cancer patients. see oncology patients<br />

cardiac disease<br />

and influenza vaccination, 252<br />

and pneumococcal disease, 327<br />

carers<br />

influenza vaccination, 255<br />

measles vaccination, 274<br />

vaccination recommendations, 170<br />

catch-up vaccination, 8, 38–64<br />

for 13vPCV and 23vPPV in children with a<br />

medical condition(s) associated with an<br />

increased risk of invasive pneumococcal<br />

disease, 59–60<br />

for 13vPCV in Indigenous children in NT,<br />

Qld, SA or WA, 57–58<br />

for 13vPCV in non-Indigenous children,<br />

and Indigenous children in the ACT,<br />

NSW, Tas and Vic, who do not have any<br />

medical condition(s) associated with an<br />

increased risk of invasive pneumococcal<br />

disease, 56<br />

children


and travellers, 119<br />

vaccines, 177–178<br />

variations from product information, 181<br />

chronic cardiac disease, and pneumococcal<br />

disease, 327<br />

chronic conditions, vaccination<br />

recommendations, 35, 165<br />

chronic inherited metabolic diseases, and<br />

influenza vaccination, 254<br />

chronic liver disease patients<br />

hepatitis A, 204<br />

hepatitis B, 222<br />

pneumococcal disease, 327<br />

chronic lung disease, and pneumococcal<br />

disease, 327<br />

chronic neurological conditions, and influenza<br />

vaccination, 253<br />

chronic renal failure, and influenza vaccination,<br />

254<br />

chronic respiratory conditions, and influenza<br />

vaccination, 253<br />

Clostridium tetani, 51, 397, 405, 463. see also<br />

tetanus<br />

CMV Immunoglobulin-VF (human), 461<br />

co-administration of vaccines, 8. see also<br />

simultaneous injections<br />

cholera, 179<br />

Haemophilus influenzae type b (Hib), 194, 203<br />

hepatitis A, 203<br />

hepatitis B, 217, 237<br />

human papillomavirus (HPV), 237<br />

influenza, 92, 251<br />

Japanese encephalitis, 263<br />

measles, 271<br />

meningococcal disease, 288, 293<br />

MMR (measles-mumps-rubella) vaccines,<br />

271, 301<br />

MMRV (measles-mumps-rubella-varicella)<br />

vaccines, 271, 301<br />

mumps, 298<br />

pneumococcal disease, 323–324, 335<br />

rotavirus, 376<br />

rubella, 388<br />

typhoid, 419<br />

varicella, 427<br />

yellow fever, 441<br />

zoster (herpes zoster), 449–450, 453<br />

cochlear implants, and pneumococcal disease,<br />

326<br />

cold chain, 25<br />

complement component disorders, and<br />

meningococcal vaccination, 291<br />

congenital cellular immunodeficiencies, and<br />

tuberculosis vaccination, 413<br />

congenital limb malformation, injection sites,<br />

74<br />

congenital rubella syndrome (CRS), 384<br />

congenital varicella syndrome, 423<br />

consent, 25–28<br />

on behalf of a child or adolescent, 26–27<br />

on behalf of an adult lacking capacity, 27<br />

evidence of, 27–28<br />

for school-based vaccination programs, 28<br />

contraindications to vaccination, 37. see also<br />

under individual disease names<br />

commonly asked questions about, 477–481<br />

false contraindications, 37, 38<br />

convulsions, persons with family history, and<br />

measles vaccination, 278<br />

correctional facility inmates, 174<br />

hepatitis A, 204, 205<br />

hepatitis B, 216, 223, 224<br />

influenza, 255<br />

correctional facility staff<br />

measles, 274<br />

vaccination recommendations, 170–171<br />

corticosteroid administration, 478. see also<br />

immunosuppressive therapy<br />

and live attenuated vaccines, 146<br />

and measles vaccination, 275–276, 277<br />

and pneumococcal vaccination, 326<br />

and rabies post-exposure prophylaxis, 362<br />

and rubella vaccination, 392<br />

and tuberculosis vaccination, 413<br />

and varicella vaccination, 326<br />

and zoster vaccination, 453<br />

Coxiella burnetii, 345, 347. see also Q fever<br />

cuddle position, 75, 77<br />

cytomegalovirus immunoglobulin, 461<br />

availability, 457<br />

D<br />

day-care workers. see early childhood<br />

education and care workers<br />

deltoid injection site, 81–82<br />

detention centre staff, vaccination<br />

recommendations, 171<br />

developmental disability, persons with<br />

hepatitis A vaccination, 204<br />

hepatitis B vaccination, 223<br />

diabetes, type 1, vaccination recommendations,<br />

165<br />

diabetes mellitus<br />

and influenza vaccination, 254<br />

and pneumococcal disease, 327<br />

diphtheria, 182–190<br />

adverse events following immunisation<br />

(AEFI), 187–188<br />

bacteriology, 182<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 11–12<br />

clinical features, 182<br />

contraindications to vaccination, 187<br />

dosage and administration, 186<br />

INDEX 505<br />

INDEX


epidemiology, 182<br />

pregnancy and breastfeeding, 135, 187<br />

public health management, 188<br />

recommendations, 186–187<br />

transport, storage and handling, 185<br />

and travellers, 117<br />

vaccines, 183–185<br />

variations from product information,<br />

188–190<br />

diphtheria antitoxin, 463<br />

diphtheria-tetanus (dT) vaccine, 184, 400, 490<br />

adverse events following immunisation<br />

(AEFI), 405<br />

catch-up vaccination schedules for those<br />

≥10 years of age, 63<br />

vaccination recommendations, 401–402,<br />

403–404<br />

diphtheria-tetanus-acellular pertussis (dTpa<br />

and DTPa) vaccines, 183, 303, 304–307,<br />

398–400, 490<br />

adverse events following immunisation<br />

(AEFI), 312–313<br />

catch-up vaccination for children, 46, 50, 51<br />

catch-up vaccination schedules for those<br />

≥10 years of age, 63<br />

dosage and administration, 401<br />

and HIV-infected persons, 159<br />

minimum acceptable age for the 1st vaccine<br />

dose, 46<br />

minimum acceptable vaccine dose<br />

intervals, 50<br />

number of vaccine doses for children, 49<br />

and pneumococcal vaccination, 335<br />

and pregnancy, 135, 310–311<br />

vaccination recommendations, 308–309,<br />

401, 403–404<br />

diphtheria-tetanus-whole-cell pertussis<br />

(DTPw) vaccine, 303, 490<br />

adverse events following immunisation<br />

(AEFI), 312<br />

diseases. see vaccine-preventable diseases<br />

distraction technique, 70<br />

documentation of vaccination, 97<br />

when incomplete, 40<br />

dosage. see also under individual disease names,<br />

valid dose determination, 41<br />

dose intervals, minimum. see minimum<br />

acceptable dose intervals for children<br />

Down syndrome, 8<br />

and influenza vaccination, 13, 252<br />

and pneumococcal disease, 327<br />

drugs, persons who inject, 36, 61<br />

hepatitis A, 198, 204, 205<br />

hepatitis B, 209, 210, 216, 222, 224, 225<br />

tetanus, 403<br />

vaccination recommendations, 175<br />

506 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

dT vaccines. see diphtheria-tetanus vaccine<br />

(dT) vaccines<br />

DTPa-containing vaccine. see diphtheriatetanus-acellular<br />

pertussis (dTpa and DTPa)<br />

vaccines<br />

dTpa-containing vaccines. see diphtheriatetanus-acellular<br />

pertussis (dTpa and DTPa)<br />

vaccines<br />

Dukoral, 177<br />

dosage and administration, 178–179<br />

vaccination recommendations, 179<br />

variations from product information, 181<br />

E<br />

early childhood education and care workers<br />

influenza, 255<br />

measles, 274<br />

pertussis, 310<br />

vaccination recommendations, 169, 170<br />

varicella, 430<br />

eculizumab treatment, and meningococcal<br />

vaccination, 291<br />

effectiveness of vaccines, 20–21<br />

efficacy of vaccines, 20–21<br />

egg allergies, 10, 131, 132–133, 481<br />

and influenza vaccination, 132, 256–257, 258<br />

and measles vaccination, 276<br />

and Q fever vaccination, 351<br />

and yellow fever vaccination, 443<br />

egg protein, in vaccines, 19, 470, 486<br />

embalmers<br />

tuberculosis, 413<br />

vaccination recommendations, 172<br />

emergency workers<br />

influenza, 255<br />

vaccination recommendations, 171<br />

Engerix-B, 160, 211, 212<br />

dosage and administration, 123, 217<br />

vaccination recommendations, 222, 227<br />

vaccination schedule, 214<br />

vaccination schedule, accelerated, 215, 216<br />

vaccine components, 469, 472<br />

enteric fever. see typhoid<br />

epiglottitis, 191<br />

erythematous skin reactions, 351<br />

Essen, 363<br />

essential services personnel, influenza<br />

vaccination, 255<br />

Europe<br />

diphtheria, 187<br />

hepatitis B, 209<br />

influenza, 245<br />

Japanese encephalitis, 261<br />

measles, 268<br />

mumps, 295<br />

poliomyelitis, 338


abies and other lyssaviruses (including<br />

<strong>Australian</strong> bat lyssavirus), 353, 354<br />

tick-borne encephalitis, 114, 121<br />

tuberculosis, 408, 409<br />

F<br />

failure of vaccines, 20–21<br />

fainting. see vasovagal episode<br />

fatal disseminated infection, and tuberculosis<br />

vaccination, 414<br />

febrile convulsions, 92–93, 94<br />

and influenza vaccination, 244, 249, 257<br />

and measles vaccination, 278, 279<br />

and pertussis vaccination, 312<br />

and varicella vaccination, 434<br />

febrile seizures, 493<br />

and measles vaccination, 272, 279<br />

and mumps vaccination, 299<br />

and pneumococcal vaccination, 334, 335<br />

and rubella vaccination, 388–389<br />

and varicella vaccination, 428<br />

Fluad, 248<br />

Fluarix, 247<br />

vaccine components, 470–472<br />

Fluvax, 92, 248<br />

adverse events following immunisation<br />

(AEFI), 249, 257<br />

vaccine components, 469–472<br />

variations from product information, 258<br />

Fluvax Junior, 92<br />

adverse events following immunisation<br />

(AEFI), 257<br />

formaldehyde, in vaccines, 19, 470, 486<br />

funeral workers, vaccination recommendations,<br />

172<br />

G<br />

Gardasil, 234, 235<br />

vaccine components, 470–472<br />

variations from product information, 242<br />

gastroenteritis, and rotavirus vaccination, 380<br />

gelatin, in vaccines, 19, 471, 485<br />

gentamicin, in vaccines, 471<br />

glossary of technical terms, 489–494<br />

glutaraldehyde, in vaccines, 471<br />

glycine, in vaccines, 485<br />

Guillain-Barré syndrome (GBS), 93<br />

and influenza vaccination, 257<br />

and meningococcal vaccination, 292, 293<br />

vaccination recommendations, 34<br />

H<br />

haematopoietic stem cell transplant (HSCT)<br />

recipients<br />

Haemophilus influenzae type b (Hib), 196<br />

hepatitis B, 222<br />

meningococcal disease, 291<br />

rotavirus, 373<br />

vaccination recommendations, 155–157,<br />

156–157<br />

zoster, 447<br />

haemodialysis patients, hepatitis B vaccination,<br />

222<br />

haemoglobinopathies<br />

and influenza vaccination, 254<br />

and pneumococcal vaccination, 326–327<br />

Haemophilus influenzae, 191, 192<br />

Haemophilus influenzae type b (Hib), 191–197<br />

adverse events following immunisation<br />

(AEFI), 196<br />

bacteriology, 191<br />

catch-up vaccination for children, 46, 49,<br />

50, 51, 54–55<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 12<br />

clinical features, 191<br />

co-administration with other vaccines, 203<br />

contraindications to vaccination, 196<br />

dosage and administration, 194<br />

epidemiology, 191–192<br />

and HIV-infected persons, 159<br />

and Indigenous children, 106–107<br />

interchangeability of vaccines, 194<br />

minimum acceptable age for the 1st vaccine<br />

dose, 46<br />

minimum acceptable vaccine dose<br />

intervals, 50<br />

number of vaccine doses for children, 49<br />

and persons with asplenia, 162, 163–164<br />

pregnancy and breastfeeding, 136, 196<br />

and preterm infants, 144<br />

public health management, 197<br />

recommendations, 195–196<br />

route of vaccine administration, 68–69<br />

transport, storage and handling, 194<br />

vaccines, 192–193<br />

variations from product information, 197<br />

Haemophilus influenzae type b–meningococcal C<br />

combination vaccine (Hib-MenCCV), 12, 14,<br />

52, 192–193, 285–286, 288. see also Haemophilus<br />

influenzae type b (Hib); meningococcal<br />

disease<br />

Hajj pilgrimage, 116, 120, 129, 290. see also<br />

Middle East<br />

HALO principle, 61–63<br />

Havrix 1440, 199<br />

dosage and administration, 125, 202<br />

Havrix Junior, 199<br />

recommended dose and schedule, 202<br />

recommended lower age limit, 127<br />

vaccine components, 469–472<br />

HBsAg-positive mothers, hepatitis B<br />

vaccination, 219<br />

INDEX 507<br />

INDEX


H-B-Vax II, 212, 217<br />

dosage and administration, 123, 211, 222<br />

vaccination schedule, 156, 214<br />

vaccine components, 469, 472<br />

healthcare workers<br />

hepatitis B, 228<br />

influenza, 255<br />

measles, 274<br />

pertussis, 310<br />

poliomyelitis, 342<br />

rubella, 389–390<br />

tuberculosis, 413<br />

vaccination recommendations, 169, 170<br />

varicella, 430<br />

hepatitis A<br />

adverse events following immunisation<br />

(AEFI), 207<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 12<br />

clinical features, 198<br />

co-administration with other vaccines, 203<br />

dosage and administration, 201–203, 202<br />

epidemiology, 198–199<br />

and HIV-infected persons, 160<br />

and Indigenous persons, 107, 203<br />

interchangeability of vaccines, 203<br />

minimum acceptable age for the 1st vaccine<br />

dose, 47<br />

minimum acceptable vaccine dose<br />

intervals, 50<br />

number of vaccine doses for children, 49<br />

pregnancy and breastfeeding, 137, 206<br />

public health management, 207<br />

recommendations, 203–206<br />

route of vaccine administration, 68–69<br />

serological testing for immunity, 206<br />

transport, storage and handling, 201<br />

and travellers, 119<br />

vaccines, 199–201<br />

variations from product information, 207<br />

virology, 198<br />

hepatitis A/hepatitis B combination vaccines,<br />

205, 217, 224<br />

hepatitis A/typhoid combination vaccines,<br />

205–206<br />

hepatitis B, 208–230<br />

adverse events following immunisation<br />

(AEFI), 227–228<br />

booster doses, 224–225<br />

catch-up vaccination guidelines for<br />

children, 51<br />

catch-up vaccination schedules for those<br />

≥10 years of age, 63–64<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 12<br />

clinical features, 208<br />

combination vaccines, 224<br />

508 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

contraindications to vaccination, 227<br />

dosage and administration, 217–218<br />

epidemiology, 209–210<br />

hepatitis B immunoglobulin (HBIG), 219<br />

higher risk groups, 220–224<br />

and HIV-infected persons, 160<br />

and Indigenous persons, 109, 221<br />

interchangeability of vaccines, 217<br />

minimum acceptable age for the 1st vaccine<br />

dose, 46<br />

minimum acceptable vaccine dose<br />

intervals, 50<br />

non-responders to vaccination, 226<br />

number of vaccine doses for children, 49<br />

pregnancy and breastfeeding, 138, 227<br />

and preterm infants, 144<br />

public health management, 228–229<br />

recommendations, 218–227<br />

route of vaccine administration, 68–69<br />

serological testing for immunity, 225<br />

transport, storage and handling, 217<br />

and travellers, 117–118<br />

vaccination schedules, 214–215<br />

vaccination schedules, accelerated, 215–216<br />

variations from product information, 230<br />

virology, 208<br />

hepatitis B immunoglobulin (HBIG), 229, 460<br />

availability, 457<br />

and HBsAg-positive mothers, 219<br />

Hepatitis B Immunoglobulin-VF, 229<br />

hepatitis C patients, hepatitis B vaccination,<br />

222<br />

herpes zoster (HZ), 423. see also zoster (herpes<br />

zoster)<br />

and varicella vaccination, 434<br />

Hiberix, 51, 193<br />

transport, storage and handling, 194<br />

variations from product information, 197<br />

Hirschsprung’s disease, and rotavirus<br />

vaccination, 380<br />

history of vaccination. see vaccination status<br />

and history<br />

HIV-infected persons. see also<br />

immunocompromised persons<br />

categories of immunocompromise, 158<br />

hepatitis B, 220, 222<br />

measles, 277<br />

rubella, 392<br />

tuberculosis, 413<br />

typhoid, 421<br />

vaccination recommendations, 158–160<br />

varicella, 431<br />

yellow fever, 444<br />

zoster, 447, 450–451, 454<br />

homeless people, influenza vaccination, 255<br />

homosexuals. see men who have sex with men<br />

HPV Register. see National HPV Vaccination<br />

Program Register (NHVPR)


HPV Vaccination Program, 234<br />

human diploid cell vaccine (HDCV), 68, 133,<br />

355, 356, 358, 359, 363, 365, 370<br />

variations from product information, 371<br />

human papillomavirus (HPV)<br />

adverse events following immunisation<br />

(AEFI), 241–242<br />

catch-up vaccination schedules, 63–64<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 13<br />

clinical features, 231–232<br />

co-administration with other vaccines, 237<br />

contraindications to vaccination, 241<br />

dosage and administration, 236–237<br />

epidemiology, 232–234<br />

female vaccination, 237–238<br />

and HIV-infected persons, 159–160<br />

interchangeability of vaccines, 237<br />

male vaccination, 238–239<br />

National HPV Vaccination Program<br />

Register (NHVPR), 101–102<br />

pregnancy and breastfeeding, 139, 240<br />

recommendations, 237–240<br />

route of vaccine administration, 68–69<br />

transport, storage and handling, 236<br />

vaccines, 234–236<br />

variations from product information, 242<br />

virology, 231<br />

human rabies immunoglobulin, 357, 362–363<br />

and post-exposure prophylaxis, 360–368<br />

and pregnancy and breastfeeding, 370<br />

hypopituitarism, vaccination<br />

recommendations, 165<br />

hypotonic-hyporesponsive episode (HHE), 93<br />

and pertussis vaccination, 312–313<br />

I<br />

immigrants. see migrants to Australia<br />

immunisation, commonly asked questions,<br />

487–488<br />

active, 18–20<br />

passive, 17, 18, 456–464<br />

immunisation handbook<br />

background, 1–2<br />

development, 3–4<br />

fundamentals of immunisation, 18–23<br />

how to use, 5–6<br />

literature search strategy, 467–468<br />

what’s new, 7–17<br />

<strong>Immunisation</strong> History Statements, 99–100<br />

immunisation registers, 85–97<br />

immunocompromised persons. see also<br />

immunocompromising conditions<br />

hepatitis B, 222<br />

human papillomavirus (HPV), 239<br />

influenza, 253–254<br />

measles, 275–276, 277<br />

rotavirus, 373, 379, 380–381<br />

rubella, 392<br />

typhoid, 421<br />

vaccination recommendations, 32, 145–165<br />

vaccination recommendations for<br />

household members of, 36, 148<br />

varicella, 430, 431<br />

yellow fever, 444<br />

zoster, 435–436, 447, 450, 451–452, 453<br />

immunocompromising conditions. see<br />

also immunocompromised persons;<br />

immunosuppressive therapy<br />

asplenia, 161–164<br />

haematopoietic stem cell transplant (HSCT)<br />

recipients, 161–164<br />

HIV-infected persons, 158–160<br />

persons with autoimmune and other<br />

chronic conditions, 165<br />

solid organ transplant (SOT) recipients,<br />

150–154<br />

immunoglobulin administration, and zoster<br />

vaccination, 454<br />

immunoglobulin recipients. see normal human<br />

immunoglobulin (NHIG) recipients<br />

immunoglobulin treatment. see normal human<br />

immunoglobulin (NHIG) treatment<br />

immunoglobulins, 456–464<br />

adverse events, 464<br />

availability, 457<br />

contraindications to use, 464<br />

for intramuscular use, 457–459<br />

normal human immunoglobulins (NHIG),<br />

456<br />

specific immunoglobulins, 456–457,<br />

459–463<br />

immunosuppressive therapy<br />

and measles vaccination, 277<br />

and rabies post-exposure prophylaxis, 362<br />

and rubella vaccination, 392<br />

and tuberculosis vaccination, 413<br />

and varicella vaccination, 431<br />

and zoster immunoglobulin, 435–436, 461<br />

and zoster vaccination, 453<br />

Imogam Rabies Pasteurized, 355<br />

Imojev, 37, 111, 260–261<br />

adverse events following immunisation<br />

(AEFI), 265–266<br />

booster doses, 264<br />

dosage and administration, 125, 262–263<br />

lower age limits for, 127<br />

pregnancy and breastfeeding, 140, 264–265<br />

impaired immunity. see immunocompromising<br />

conditions<br />

impaired immunity, persons with. see<br />

immunocompromised persons<br />

INDEX 509<br />

INDEX


inactivated poliomyelitis vaccine (IPV),<br />

68–69, 118, 138, 156, 194, 339, 341. see also<br />

poliomyelitis<br />

Indigenous community workers, vaccination<br />

recommendations, 172<br />

Indigenous persons. see Aboriginal and Torres<br />

Strait Islander people<br />

indurated lesions, 352<br />

Infanrix hexa, 51, 67, 183, 193, 195, 211, 305,<br />

339–340, 342, 399<br />

dosage and administration, 214, 217<br />

transport, storage and handling, 185, 194,<br />

217, 307, 341, 401<br />

vaccination schedule, 214<br />

vaccine components, 470–472<br />

variations from product information, 188,<br />

190, 197, 230, 314, 316, 343, 405, 407<br />

Infanrix IPV, 184, 305, 340, 342, 399<br />

vaccine components, 469–472<br />

variations from product information, 188,<br />

190, 314, 316, 343–344, 406, 407<br />

infants<br />

cholera, 186<br />

diphtheria, 186<br />

Haemophilus influenzae type b (Hib), 195<br />

hepatitis B, 212, 214–215, 218–220, 219<br />

influenza, 251<br />

injection sites, 73<br />

measles, post-exposure prophylaxis, 281<br />

measles, vaccination, 271–272<br />

meningococcal disease, 290<br />

multiple vaccine injections, 83<br />

mumps, 299<br />

pertussis, 307–308, 309–310<br />

poliomyelitis, 342<br />

positioning for vaccination, 75–76, 77<br />

rabies, 356–357<br />

rotavirus, 377–378, 380–381<br />

rubella, 388<br />

tetanus, 401<br />

tuberculosis, 410, 412, 414<br />

vaccination recommendations for parents,<br />

grandparents and carers, 36, 309<br />

yellow fever, 443, 445<br />

zoster immunoglobulin, 435–436<br />

influenza<br />

adverse events following immunisation<br />

(AEFI), 257–258<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 13<br />

clinical features, 243–244<br />

co-administration with other vaccines, 251<br />

contraindications to vaccination, 256<br />

dosage and administration, 250–251<br />

and egg allergies, 256–257<br />

epidemiology, 244–246<br />

higher risk groups, 252–256<br />

510 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

and HIV-infected persons, 160<br />

and immunocompromised persons, 145<br />

and Indigenous persons, 108, 109–110, 254<br />

national notification and hospitalisation<br />

rates, 245<br />

and persons with asplenia, 162, 163–164<br />

precautions, 256–257<br />

pregnancy and breastfeeding, 135, 256<br />

and preterm infants, 144<br />

public health management, 258<br />

recommendations, 251–256<br />

route of vaccine administration, 68–69<br />

transport, storage and handling, 250<br />

and travellers, 118<br />

vaccination and egg allergies, 132<br />

vaccines, 246–250<br />

variations from product information, 258<br />

virology, 243<br />

influenza pandemic, 245–246<br />

Influvac, 247<br />

vaccine components, 470–472<br />

Influvac Junior, 247<br />

injecting drug users. see drugs, persons who<br />

inject<br />

injection of vaccines, 66–67<br />

injection site abscess<br />

and Q fever vaccination, 351<br />

and tuberculosis vaccination, 414<br />

injection site nodules, 91<br />

injection site reactions, 63, 74<br />

and human papillomavirus (HPV)<br />

vaccination, 241<br />

and Japanese encephalitis vaccination,<br />

265–266<br />

and pneumococcal vaccination, 323<br />

and tetanus vaccination, 404–405<br />

and varicella vaccination, 433<br />

and zoster vaccination, 448, 451, 452, 454<br />

injection sites<br />

adolescents, 74<br />

adults, 74<br />

breast cancer or lymphoedema patients, 74<br />

children, 73–74<br />

children in spica casts, 74<br />

children with congenital limb<br />

malformation, 74<br />

identification of, 79–82<br />

infants, 73<br />

multiple injections, 83–84<br />

injection techniques<br />

intradermal injection, 72<br />

intramuscular injection, 71–73<br />

subcutaneous injection, 72<br />

inmates of correctional facilities. see<br />

correctional facility inmates<br />

Intanza 9 mg, 68, 72, 248, 249<br />

Intanza 15 mg, 68, 72, 248, 249


interferon-gamma release assays (IGRAs), 410<br />

International Certificate of Vaccination or<br />

Prophylaxis against yellow fever, 440,<br />

441–442<br />

international travel. see travellers<br />

interruption to a vaccination, 73, 376, 383<br />

intracranial shunts, and pneumococcal disease,<br />

326<br />

intradermal injection technique, 72<br />

intradermal vaccines, 68–69<br />

intramuscular (IM) injection technique, 71<br />

intramuscular (IM) injection vaccines, 68–69<br />

intussusception (IS), and rotavirus vaccination,<br />

16, 22, 93, 374–375, 379, 381–382<br />

invasive pneumococcal disease (IPD). see<br />

pneumococcal disease<br />

IPOL, 69, 339, 341<br />

adverse events following immunisation<br />

(AEFI), 343<br />

contraindications to vaccination, 343<br />

dosage and administration, 124, 341<br />

pregnancy and breastfeeding, 343<br />

vaccination recommendations, 342<br />

J<br />

Japanese encephalitis<br />

adverse events following immunisation<br />

(AEFI), 265–266<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 13<br />

clinical features, 259<br />

co-administration with other vaccines, 263<br />

contraindications to vaccination, 265<br />

dosage and administration, 262–263<br />

epidemiology, 259–260<br />

and Indigenous persons, 111<br />

precautions, 265<br />

pregnancy and breastfeeding, 138, 140,<br />

264–265<br />

public health management, 266<br />

recommendations, 263–264<br />

transport, storage and handling, 262<br />

and travellers, 119<br />

vaccines, 260–262<br />

variations from product information, 266<br />

virology, 259<br />

JEspect, 68, 111, 260, 261–262<br />

adverse events following immunisation<br />

(AEFI), 266<br />

dosage and administration, 125, 262–263<br />

lower age limits for, 127–128<br />

precautions, 265<br />

pregnancy and breastfeeding, 138, 265<br />

vaccination recommendations, 263–264<br />

variations from product information, 266<br />

K<br />

kanamycin, in vaccines, 471<br />

keloid formation, and tuberculosis vaccination,<br />

411, 414<br />

L<br />

laboratory personnel<br />

Japanese encephalitis, 264<br />

meningococcal disease, 290, 291<br />

poliomyelitis, 342<br />

vaccination recommendations, 171<br />

lactose, in vaccines, 19, 485<br />

leprosy, and tuberculosis vaccination, 412<br />

leukaemia. see immunocompromised persons<br />

lifestyle vaccination requirements, 36<br />

limb swelling, and pertussis vaccination, 308,<br />

312, 491<br />

Liquid PedvaxHIB, 51, 55, 193<br />

variations from product information, 197<br />

live attenuated vaccines, 32, 37. see also<br />

Japanese encephalitis; MMR (measlesmumps-rubella)<br />

vaccine; MMRV (measlesmumps-rubella-varicella)<br />

vaccine; rotavirus;<br />

tuberculosis; typhoid; varicella; yellow fever;<br />

zoster (herpes zoster)<br />

and anaesthesia or surgery, 168<br />

and autoimmune conditions, 165<br />

and blood products, 150, 166<br />

and corticosteroids, 165<br />

and haematopoietic stem cell transplant<br />

(HSCT) recipients, 155<br />

and HIV-infected persons, 158–159<br />

and immunocompromised persons, 32,<br />

35–36, 142, 145, 146, 147–148<br />

and immunoglobulins, 150, 463–464<br />

and migrants to Australia, 174<br />

and normal human immunoglobulin<br />

(NHIG) recipients, 166<br />

and pregnancy, 33, 34, 133–134, 142<br />

livestock workers. see animals, persons<br />

working with<br />

local adverse event. see injection site reactions<br />

long-term care facility workers<br />

influenza, 255<br />

measles, 274<br />

vaccination recommendations, 170<br />

varicella, 430<br />

lymphoedema patients, injection sites, 74<br />

lymphoid system malignancies, and<br />

tuberculosis vaccination, 413<br />

lymphoma. see immunocompromised persons<br />

Lyssavirus, 353. see also rabies and other<br />

lyssaviruses (including <strong>Australian</strong> bat<br />

lyssavirus)<br />

lyssaviruses. see rabies and other lyssaviruses<br />

(including <strong>Australian</strong> bat lyssavirus)<br />

INDEX 511<br />

INDEX


M<br />

malabsorption syndromes, and rotavirus<br />

vaccination, 380<br />

malnutrition, and tuberculosis vaccination, 413<br />

mannitol, in vaccines, 19, 471, 485<br />

Mantoux test. see tuberculin skin test<br />

manufacturing residuals, in vaccines, 486<br />

measles<br />

adverse events following immunisation<br />

(AEFI), 278–280<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 13–14<br />

clinical features, 267<br />

co-administration with other vaccines, 271<br />

contraindications to vaccination, 275–276<br />

dosage and administration, 271<br />

epidemiology, 267–268<br />

global elimination, 268<br />

higher risk groups, 274–275<br />

and other live attenuated parenteral<br />

vaccines, 276<br />

precautions, 276–278<br />

pregnancy and breastfeeding, 275<br />

public health management, 280–281<br />

recommendations, 271–275<br />

serological testing for immunity, 274–275<br />

transport, storage and handling of vaccines,<br />

270<br />

and travellers, 118<br />

vaccines, 269–270<br />

variations from product information, 282<br />

virology, 267<br />

measles control campaigns, 268, 296, 384<br />

measles-mumps-rubella vaccine. see MMR<br />

(measles-mumps-rubella) vaccine<br />

measles-mumps-rubella-varicella vaccine. see<br />

MMRV (measles-mumps-rubella-varicella)<br />

vaccine<br />

meat industry workers. see animals, persons<br />

working with<br />

Medicare Online, 98<br />

men who have sex with men, 174<br />

hepatitis A, 204, 205<br />

hepatitis B, 221, 224<br />

human papillomavirus (HPV), 232, 239<br />

Menactra, 285–286<br />

adverse events following immunisation<br />

(AEFI), 292<br />

dosage and administration, 125<br />

lower age limits for, 127<br />

variations from product information, 293<br />

Mencevax ACWY, 287<br />

transport, storage and handling, 288<br />

Meningitec, 285<br />

vaccine components, 470<br />

variations from product information, 293<br />

512 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

meningitis. see Haemophilus influenzae type b<br />

(Hib); meningococcal disease<br />

meningococcal C conjugate vaccines<br />

(MenCCV), 284, 285, 289–290. see also<br />

meningococcal disease<br />

dosage and administration, 288<br />

minimum acceptable age for the 1st vaccine<br />

dose, 47<br />

variations from product information, 293<br />

meningococcal disease, 283–294<br />

adverse events following immunisation<br />

(AEFI), 292<br />

bacteriology, 283<br />

catch-up vaccination guidelines for<br />

children, 52<br />

catch-up vaccination schedules for those<br />

≥10 years of age, 63–64<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 14<br />

clinical features, 283<br />

contraindications to vaccination, 292<br />

dosage and administration, 288–289<br />

epidemiology, 284<br />

and haematopoietic stem cell transplant<br />

(HSCT) recipients, 157<br />

higher risk groups, 289–291<br />

interchangeability of vaccines, 289<br />

minimum acceptable vaccine dose<br />

intervals, 50<br />

number of vaccine doses for children, 49<br />

and persons with asplenia, 161–162,<br />

163–164<br />

pregnancy and breastfeeding, 136, 291–292<br />

public health management, 292–293<br />

recommendations, 289–291<br />

and solid organ transplant (SOT) recipients,<br />

153<br />

transport, storage and handling, 287–288<br />

and travellers, 120<br />

vaccines, 69, 285–287<br />

variations from vaccine product<br />

information, 293–294<br />

Menitorix, 51, 52, 55, 193, 285<br />

transport, storage and handling, 194, 288<br />

Menjugate Syringe, 285<br />

transport, storage and handling, 287<br />

vaccine components, 469<br />

variations from product information, 293<br />

Menomune, 287<br />

dosage and administration, 126<br />

lower age limits for, 127<br />

transport, storage and handling, 288<br />

Menveo, 286<br />

dosage and administration, 125<br />

lower age limits for, 127<br />

transport, storage and handling, 288<br />

variations from product information, 293


mercury, in vaccines. see thiomersal, in vaccines<br />

Mérieux Inactivated Rabies Vaccine, 133, 355.<br />

see also human diploid cell vaccine (HDCV)<br />

dosage and administration, 126<br />

metabolic diseases<br />

and rotavirus vaccination, 380<br />

vaccination recommendations, 165<br />

Middle East<br />

hepatitis B, 209, 221<br />

meningococcal disease, 116, 120, 290<br />

pilgrimage to Mecca (Hajj and Umra), 116,<br />

120, 129, 290<br />

migrants to Australia, 173–174<br />

from hepatitis B endemic countries, 12,<br />

209, 221<br />

immunisation schedules for other<br />

countries, 39<br />

and travel, 127<br />

tuberculosis, 412<br />

typhoid, 417<br />

military personnel. see armed forces<br />

minimum acceptable age for the 1st dose of<br />

vaccination, 8, 41, 46–48<br />

minimum acceptable dose intervals for<br />

children, 41, 42, 50<br />

MMR (measles-mumps-rubella) vaccines,<br />

269–270, 296–297, 386, 425<br />

and adults and adolescents, 273<br />

adverse events following immunisation<br />

(AEFI), 278–280, 301, 394<br />

catch-up vaccination guidelines for<br />

children, 52<br />

catch-up vaccination schedules for those<br />

≥10 years of age, 63–64<br />

and children, 272–273, 299<br />

co-administration with other vaccines,<br />

271, 301<br />

contraindications to vaccination, 392–393<br />

dosage and administration, 271, 387–388<br />

and HIV-infected persons, 158–159<br />

and immunocompromised persons, 147<br />

and infants, 274, 299<br />

interchangeability of vaccines, 271<br />

minimum acceptable age for the 1st vaccine<br />

dose, 47<br />

minimum acceptable vaccine dose<br />

intervals, 50<br />

number of vaccine doses for children, 49<br />

precautions, 276–278, 393–394<br />

pregnancy and breastfeeding, 140, 275, 300,<br />

391–392<br />

recommended intervals between<br />

immunoglobulins or blood products and<br />

the vaccine, 167<br />

and tuberculosis vaccination, 414<br />

vaccination and egg allergies, 132–133<br />

vaccination of suspected rubella contacts,<br />

394–395<br />

vaccination recommendations, 388–391<br />

vaccination schedule, 273<br />

variations from product information, 282,<br />

396<br />

and yellow fever vaccination, 444<br />

and zoster vaccination, 450<br />

M-M-R II, 68, 269, 297, 386<br />

dosage and administration, 124, 271, 298,<br />

387<br />

transport, storage and handling, 270, 298,<br />

387<br />

variations from product information, 282<br />

MMRV (measles-mumps-rubella-varicella)<br />

vaccines, 68–69, 270, 296–297, 386, 424, 425,<br />

426<br />

and adults and adolescents, 273, 299–300,<br />

428–429<br />

adverse events following immunisation<br />

(AEFI), 278–280, 301, 394<br />

catch-up vaccination guidelines for<br />

children, 52<br />

and children, 272–273, 299, 428<br />

co-administration with other vaccines,<br />

271, 301<br />

dosage and administration, 271, 387–388,<br />

427<br />

and immunocompromised persons, 147<br />

precautions, 276–278, 393–394<br />

pregnancy and breastfeeding, 140, 275, 300,<br />

391–392<br />

recommended intervals between<br />

immunoglobulins or blood products and<br />

the vaccine, 167<br />

and tuberculosis vaccination, 414<br />

vaccination of suspected rubella contacts,<br />

394–395<br />

vaccination recommendations, 388–391<br />

vaccination schedule, 273<br />

variations from product information, 282,<br />

396, 437–438<br />

and varicella vaccination, 430–431<br />

and yellow fever vaccination, 444<br />

Modified Essen, 363<br />

monosodium glutamate (MSG), in vaccines,<br />

471, 485<br />

Morbillivirus, 267. see also measles<br />

multi-dose vials, 67<br />

multiple sclerosis, vaccination<br />

recommendations, 165<br />

mumps, 295–301<br />

adverse events following immunisation<br />

(AEFI), 301<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 13–14<br />

clinical features, 295<br />

INDEX 513<br />

INDEX


co-administration with other vaccines, 298<br />

contraindications to vaccination, 300<br />

dosage and administration, 298<br />

epidemiology, 295–296<br />

interchangeability of vaccines, 298<br />

precautions, 300–301<br />

pregnancy and breastfeeding, 300<br />

public health management, 301<br />

recommended doses and schedules,<br />

299–300<br />

serological testing for immunity, 300<br />

transport, storage and handling, 298<br />

and travellers, 118<br />

vaccines, 296<br />

variations from product information, 301<br />

virology, 295<br />

Mycobacterium africanum, 408. see also<br />

tuberculosis<br />

Mycobacterium bovis, 408, 409. see also<br />

tuberculosis<br />

Mycobacterium canetti, 408. see also tuberculosis<br />

Mycobacterium leprae, 410, 412. see also<br />

tuberculosis<br />

Mycobacterium microti, 408. see also tuberculosis<br />

Mycobacterium tuberculosis, 408. see also<br />

tuberculosis<br />

Mycoplasma pneumoniae, 302. see also<br />

tuberculosis<br />

N<br />

National Centre for <strong>Immunisation</strong> Research<br />

and Surveillance of Vaccine Preventable<br />

Diseases (NCIRS), 3, 27, 467, 467–468, 488,<br />

498<br />

National HPV Vaccination Program Register<br />

(NHVPR), 9, 40, 101–102, 174<br />

National <strong>Immunisation</strong> Program (NIP), 2, 3,<br />

31, 67<br />

catch-up vaccination, 38–39<br />

catch-up worksheet for children


Northern Territory<br />

ACIR reporting, 98<br />

adverse events following immunisation<br />

(AEFI) notification, 96<br />

age of consent, 26<br />

<strong>Australian</strong> Red Cross Blood Service, 457<br />

government health authority and<br />

communicable disease control contact<br />

details, 465–466<br />

hepatitis A, 105, 107, 199, 201, 203<br />

hepatitis B, 109, 210<br />

HPV reporting, 101<br />

influenza, 251<br />

NT <strong>Immunisation</strong> Register, 103<br />

pneumococcal disease, 15, 52, 57–58, 105,<br />

108, 110, 318, 321, 324, 329, 330<br />

tuberculosis, 106, 107, 408<br />

nursing home staff<br />

influenza, 255<br />

vaccination recommendations, 170<br />

O<br />

obese persons, 72<br />

influenza, 252<br />

occupational health and safety issues, 65<br />

administration of vaccines, 65<br />

occupational risks, 2, 11, 29, 36, 61, 62, 115,<br />

130, 169–173, 371. see also under individual<br />

occupations<br />

hepatitis A, 199, 204, 205<br />

hepatitis B, 223–224, 225, 460<br />

measles, 274<br />

pertussis, 310<br />

Q fever, 346, 348<br />

rabies and other lyssaviruses (including<br />

<strong>Australian</strong> bat lyssavirus), 371<br />

rubella, 396<br />

tuberculosis, 413<br />

varicella, 430<br />

Oceania, 113, 118, 439. see also Pacific Islands<br />

oncology patients, vaccination<br />

recommendations, 148–150<br />

OPV, 53, 339, 341, 376<br />

oral live attenuated typhoid vaccine. see also<br />

Vivotif Oral<br />

adverse events following immunisation<br />

(AEFI), 422<br />

contraindications to vaccination, 421<br />

dosage and administration, 418–419<br />

precautions, 421–422<br />

pregnancy and breastfeeding, 421<br />

transport, storage and handling, 418<br />

oral poliomyelitis vaccine. see OPV<br />

oral vaccines, 37, 68–69, 73, 123–126. see also<br />

under rotavirus, typhoid and cholera<br />

oropharyngeal cancer, 233<br />

otitis media, 108, 191, 244, 267, 317, 321. see also<br />

pneumococcal disease<br />

P<br />

Pacific Islands. see also Oceania<br />

hepatitis B, 209, 221<br />

rabies and other lyssaviruses (including<br />

<strong>Australian</strong> bat lyssavirus), 354<br />

rubella, 390<br />

tuberculosis, 408<br />

palivizumab, 462<br />

Papua New Guinea<br />

Japanese encephalitis, 113–114, 119, 259, 263<br />

rabies and other lyssaviruses (including<br />

<strong>Australian</strong> bat lyssavirus), 354<br />

typhoid, 122, 416<br />

paracetamol, 85, 91, 292<br />

and fits, 478<br />

measles, 278, 279<br />

varicella, 434<br />

paralytic polio. see poliomyelitis<br />

parenteral Vi polysaccharide vaccines, 418. see<br />

also typhoid, vaccines<br />

adverse events following immunisation<br />

(AEFI), 422<br />

contraindications to vaccination, 421<br />

dosage and administration, 419<br />

pregnancy and breastfeeding, 421<br />

vaccination recommendations, 420<br />

passive immunisation, 18, 456–464. see also<br />

immunoglobulins<br />

availability of immunoglobulins, 457<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 17<br />

normal human immunoglobulin for<br />

intramuscular use, 457–459<br />

potential interactions with vaccines,<br />

463–464<br />

specific immunoglobulins, 459–463<br />

transport, storage and handling, 457<br />

Pediacel, 51, 184, 193, 305–306, 340, 399<br />

variations from product information, 189,<br />

197, 315, 344, 406<br />

pertussis, 302–316<br />

adverse events following immunisation<br />

(AEFI), 312–313<br />

bacteriology, 302<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 11–12<br />

clinical features, 302<br />

contraindications to vaccination, 312<br />

dosage and administration, 307<br />

interval between vaccines, 310<br />

pregnancy and breastfeeding, 310–311<br />

public health management, 313–314<br />

recommendations, 307–310<br />

transport, storage and handling, 307<br />

and travellers, 117<br />

vaccination of contacts of cases, 314<br />

vaccines, 304–307<br />

INDEX 515<br />

INDEX


variations from product information,<br />

314–316<br />

Pharmaceutical Benefits Advisory Committee<br />

(PBAC), 3<br />

Pharmaceutical Benefits Scheme (PBS), 3<br />

phenol, in vaccines, 471, 484<br />

phenoxyethanol, in vaccines, 471, 484<br />

pilgrims attending Hajj in Saudi Arabia, 116,<br />

120, 129, 290<br />

plumbers, vaccination recommendations, 172<br />

pneumococcal conjugate vaccines (PCV). see<br />

10vPCV (10-valent pneumococcal conjugate<br />

vaccine); 13vPCV (13-valent pneumococcal<br />

conjugate vaccine)<br />

pneumococcal disease, 317–337<br />

adverse events following immunisation<br />

(AEFI), 335–336<br />

bacteriology, 317<br />

catch-up vaccination for children, 46, 49, 50,<br />

52–53, 57–58, 59–60, 63<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 14–15<br />

clinical features, 317–318<br />

contraindications to vaccination, 334<br />

dosage and administration, 323–324<br />

epidemiology, 318–319<br />

and HIV-infected persons, 160<br />

and immunocompromised persons, 145<br />

immunocompromising conditions with<br />

increased risk of, 326–327<br />

and Indigenous persons, 108, 110–111<br />

and persons with asplenia, 161, 163–164<br />

precautions, 334<br />

pregnancy and breastfeeding, 334<br />

and preterm infants, 143–144<br />

recommendations, 324–333<br />

transport, storage and handling, 323<br />

and travellers, 118<br />

vaccines, 319–323<br />

variations from product information,<br />

336–337<br />

pneumococcal polysaccharide vaccine.<br />

see 23vPPV (23-valent pneumococcal<br />

polysaccharide vaccine)<br />

pneumonia. see Haemophilus influenzae type b<br />

(Hib); pneumococcal disease<br />

Pneumovax 23, 322, 449<br />

catch-up schedule, 59–60<br />

dosage and administration, 124<br />

vaccine components, 471<br />

variations from product information, 337<br />

police, vaccination recommendations, 171<br />

poliomyelitis, 338–344, 348–350<br />

adverse events following immunisation<br />

(AEFI), 343<br />

516 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

catch-up vaccination for children, 46, 49,<br />

50, 53<br />

catch-up vaccination schedules for those<br />

≥10 years of age, 63–64<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 15<br />

clinical features, 338<br />

contraindications to vaccination, 343<br />

dosage and administration, 341<br />

epidemiology, 338–339<br />

interchangeability of vaccines, 341<br />

minimum acceptable age for the 1st vaccine<br />

dose, 46<br />

minimum acceptable vaccine dose<br />

intervals, 50<br />

number of vaccine doses for children, 49<br />

pregnancy and breastfeeding, 343<br />

pre-vaccination testing, 348–349<br />

public health management, 343<br />

recommendations, 342<br />

transport, storage and handling, 341<br />

and travellers, 118<br />

variations from product information,<br />

343–344<br />

virology, 338<br />

poliovirus, 338<br />

polymyxin, in vaccines, 19, 472, 486<br />

polysorbate, in vaccines, 19, 472, 485<br />

pork industry workers. see animals, persons<br />

working with<br />

positioning for vaccination<br />

children, 77–78<br />

infants, 75–76<br />

older children, adolescents and adults, 78<br />

post-exposure prophylaxis<br />

hepatitis B, 229<br />

measles, 281<br />

varicella, 437<br />

post-herpetic neuralgia (PHN), 446<br />

post-vaccination procedures<br />

adverse events following immunisation<br />

(AEFI), 85–97<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 9<br />

documentation of vaccination, 97<br />

immediate after-care, 85<br />

immunisation registers, 98–103<br />

poultry workers. see animals, persons working<br />

with<br />

precautions to vaccination, 5, 22, 28, 130, 477.<br />

see also under individual disease names<br />

pregnancy<br />

and cholera vaccination, 180<br />

contact with persons who received live<br />

vaccines, 134, 142<br />

and diphtheria vaccination, 187


and Haemophilus influenzae type b (Hib)<br />

vaccination, 196<br />

and hepatitis A vaccination, 206<br />

and human papillomavirus (HPV)<br />

vaccination, 240<br />

and immunosuppressive therapy, 142<br />

and influenza vaccination, 252, 256<br />

and Japanese encephalitis vaccination,<br />

264–265<br />

and measles, 267<br />

and measles vaccination, 275<br />

and meningococcal vaccination, 291–292<br />

and mumps, 295<br />

and mumps vaccination, 300<br />

and pertussis vaccination, 303–304, 308,<br />

309, 310–311<br />

and pneumococcal vaccination, 334<br />

and Q fever vaccination, 350–351<br />

and rabies vaccination, 370<br />

and rotavirus vaccination, 379<br />

and rubella, 384, 395–396<br />

and rubella vaccination, 385, 391–392,<br />

392–393<br />

and tetanus vaccination, 402<br />

and tuberculosis vaccination, 413<br />

vaccination recommendations, 33–34,<br />

133–144, 135–141<br />

and varicella vaccination, 430–431, 432<br />

and yellow fever vaccination, 443, 445<br />

and zoster immunoglobulin, 435–436<br />

and zoster vaccination, 453<br />

premature infants. see preterm infants<br />

preparation for vaccine administration, 65, 70<br />

preparation of vaccines, 66–67<br />

pre-school workers. see early childhood<br />

education and care workers<br />

preservatives, in vaccines, 484<br />

preterm infants, 34, 38, 143–144, 475<br />

Haemophilus influenzae type b (Hib), 51,<br />

144, 195<br />

hepatitis B, 51, 144, 219<br />

influenza, 144<br />

needle size, length and angle, 72<br />

pneumococcal disease, 43, 53, 143–144, 327<br />

rotavirus, 144, 378–379<br />

pre-vaccination procedures, 24–64<br />

anaphylaxis response kit, 24<br />

catch-up, 38–64<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 8<br />

effective cold chain, 24–25<br />

screening, 28–38<br />

valid consent, 25–28<br />

pre-vaccination screening, 28–38<br />

checklist, 30–31<br />

responses to relevant conditions or<br />

circumstances, 31<br />

Prevenar 13, 320<br />

catch-up schedule, 56, 57–58, 59–60<br />

dosage and administration, 124, 194, 323<br />

vaccine components, 470–472<br />

variations from product information, 336<br />

Priorix, 68, 269–270, 297, 386<br />

dosage and administration, 124, 271, 298,<br />

387<br />

transport, storage and handling, 270, 298,<br />

387<br />

vaccine components, 471–472<br />

variations from product information, 282<br />

Priorix-tetra, 269, 270, 297, 386–387, 425–426<br />

dosage and administration, 298, 387, 427<br />

seroresponse assessment in study, 269<br />

transport, storage and handling, 270, 298,<br />

387, 427<br />

vaccine components, 471–472<br />

variations from product information, 282,<br />

437<br />

prisoners and staff. see correctional facility<br />

inmates; correctional facility staff<br />

procedures for vaccination, 24–103<br />

administration of vaccines, 65–84<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 8–9<br />

post-vaccination, 85–103<br />

pre-vaccination, 24–64<br />

product information for vaccines. see vaccine<br />

product information<br />

prone position, 75–76, 78<br />

ProQuad, 68, 270, 297, 387, 425–426<br />

adverse events following immunisation<br />

(AEFI), 279, 434<br />

seroresponse assessment in study, 269<br />

transport, storage and handling, 270, 298,<br />

387, 427<br />

vaccine components, 469, 471–472<br />

variations from product information, 282,<br />

437<br />

public health management, 5, 8. see also under<br />

individual disease names<br />

purified chick embryo cell vaccine (PCECV),<br />

69, 71, 133, 355, 356, 358, 363, 370<br />

variations from product information, 371<br />

Q<br />

Q fever, 345–352<br />

adverse events following immunisation<br />

(AEFI), 351–352<br />

<strong>Australian</strong> Q Fever Register, 102<br />

bacteriology, 345<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 15<br />

clinical features, 345<br />

contraindications to vaccination, 351<br />

dosage and administration, 348<br />

INDEX 517<br />

INDEX


and egg allergies, 132–133<br />

epidemiology, 345–346<br />

precautions, 351<br />

pregnancy and breastfeeding, 137, 350–351<br />

public health management, 352<br />

recommendations, 348–350<br />

route of vaccine administration, 68–69<br />

skin test, 68–69<br />

transport, storage and handling, 347<br />

vaccine, 347<br />

variations from product information, 352<br />

Q fever fatigue syndrome (QFS), 345, 351<br />

Quadracel, 184, 306, 340, 399<br />

vaccine components, 469–472<br />

variations from product information, 188,<br />

315, 344, 406<br />

quadrivalent meningococcal conjugate<br />

vaccines (4vMenCV), 14, 69, 120, 153, 157,<br />

171, 285–286. see also meningococcal disease<br />

adverse events following immunisation<br />

(AEFI), 292<br />

dosage and administration, 288<br />

vaccination recommendations, 290–291<br />

variations from product information,<br />

293–294<br />

quadrivalent meningococcal polysaccharide<br />

vaccines (4vMenPV), 14, 20, 68, 71, 120, 285,<br />

287, 290–291. see also meningococcal disease<br />

adverse events following immunisation<br />

(AEFI), 292<br />

dosage and administration, 288<br />

lower age limits for, 127<br />

and pregnancy, 136<br />

vaccination recommendations, 290–291<br />

variations from product information, 293<br />

Queensland<br />

ACIR reporting, 98<br />

adverse events following immunisation<br />

(AEFI) notification, 95, 96<br />

age of consent, 26<br />

<strong>Australian</strong> Red Cross Blood Service, 457<br />

cholera, 176<br />

government health authority and<br />

communicable disease control contact<br />

details, 465–466<br />

hepatitis A, 105, 107, 199, 201, 203, 204<br />

HPV reporting, 101<br />

influenza, 251<br />

Japanese encephalitis, 259<br />

pneumococcal disease, 15, 52, 57, 57–58,<br />

105, 108, 110, 318, 324<br />

Q fever, 346<br />

tuberculosis, 106, 408<br />

Vaccine Information and Vaccine<br />

Administration System (VIVAS), 102–103<br />

yellow fever, 439<br />

518 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

Q-Vax, 132, 347<br />

contraindications to vaccination, 351<br />

variations from product information, 352<br />

Q-Vax Skin Test, 347<br />

recommendations, 348–349<br />

skin test process and interpretation,<br />

348–350<br />

transport, storage and handling, 347<br />

R<br />

rabies and other lyssaviruses (including<br />

<strong>Australian</strong> bat lyssavirus), 68–69, 353–371<br />

adverse events following immunisation<br />

(AEFI), 370<br />

booster algorithm, 369<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 15–16<br />

clinical features, 353<br />

contraindications to vaccination, 370<br />

dosage and administration, 256–358<br />

and egg allergies, 132–133<br />

epidemiology, 354<br />

exposure categories, 360<br />

interchangeability of vaccines, 358<br />

post-exposure prophylaxis, 357, 360–368,<br />

366, 367<br />

pre-exposure prophylaxis, 356–357, 359–360<br />

pregnancy and breastfeeding, 138, 370<br />

rabies immunoglobulin, 355–356<br />

recommendations, 358–370<br />

serological testing for immunity, 369<br />

transport, storage and handling, 256<br />

and travellers, 120<br />

vaccines, 355<br />

virology, 353<br />

rabies immunoglobulin (HRIG), 357, 457,<br />

460. see also rabies and other lyssaviruses<br />

(including <strong>Australian</strong> bat lyssavirus)<br />

Rabipur Inactivated Rabies Virus Vaccine, 71,<br />

133, 355. see also purified chick embryo cell<br />

vaccine (PCECV)<br />

dosage and administration, 126<br />

radiation therapy, persons receiving. see<br />

immunosuppressive therapy<br />

rash<br />

and varicella vaccination, 433–434<br />

zoster (herpes zoster), 446<br />

and zoster vaccination, 454<br />

recommendation changes in the <strong>10th</strong> edition of<br />

the handbook, 8–17<br />

reconstitution of vaccines, 66–67<br />

refugees. see migrants to Australia<br />

registers for immunisation, 85–97<br />

regurgitation of a vaccine, 73, 376, 383<br />

renal impaired patients, hepatitis B vaccination,<br />

222<br />

respiratory papillomatosis, 234


espiratory syncytial virus monoclonal<br />

antibody, 457, 462<br />

Reye syndrome, and varicella vaccination, 438<br />

rheumatoid arthritis, vaccination<br />

recommendations, 165<br />

risks and benefits of vaccines, 27<br />

Rotarix, 374–375<br />

adverse events following immunisation<br />

(AEFI), 381–382<br />

dosage and administration, 50, 53, 376<br />

vaccination recommendations, 377–379<br />

variations from product information, 383<br />

RotaShield, 374–375<br />

RotaTeq, 374–375<br />

adverse events following immunisation<br />

(AEFI), 381–382<br />

dosage and administration, 50, 53, 376<br />

precautions, 380<br />

vaccination recommendations, 377–379<br />

vaccine components, 472<br />

variations from product information, 383<br />

rotavirus, 372–383<br />

adverse events following immunisation<br />

(AEFI), 381–382<br />

catch-up vaccination guidelines for<br />

children, 53<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 16<br />

clinical features, 372<br />

co-administration with other vaccines, 376<br />

contraindications to vaccination, 379<br />

dosage and administration, 47, 49, 50, 376<br />

epidemiology, 372–373<br />

and HIV-infected persons, 158<br />

and immunocompromised persons, 147<br />

interchangeability of vaccines, 377<br />

precautions, 380–381<br />

pregnancy and breastfeeding, 140, 379<br />

and preterm infants, 144<br />

and recipients of normal human<br />

immunoglobulin and other blood<br />

products, 166<br />

recommendations, 377–379<br />

route of vaccine administration, 68–69<br />

transport, storage and handling, 376<br />

vaccines, 374–375<br />

variations from product information, 383<br />

virology, 372<br />

rubella, 384–396<br />

adverse events following immunisation<br />

(AEFI), 394<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 13–14<br />

clinical features, 384<br />

co-administration with other vaccines, 388<br />

contraindications to vaccination, 392–393<br />

dosage and administration, 387–388<br />

epidemiology, 384–385<br />

and Indigenous persons, 111<br />

interchangeability of vaccines, 388<br />

precautions, 393–394<br />

pregnancy and breastfeeding, 391–392,<br />

392–393, 395–396<br />

public health management, 394–396<br />

recommendations, 388–391<br />

serological testing for immunity, 390–391,<br />

395<br />

transport, storage and handling, 387<br />

and travellers, 118<br />

vaccines, 386–387<br />

variations from product information, 396<br />

virology, 384<br />

and women of child-baring age, 390, 391<br />

Rubivirus, 384. see also rubella<br />

S<br />

safety of vaccines, 21–23<br />

salicylate therapy. see aspirin therapy<br />

Salmonella enterica subspecies enterica serovar<br />

Typhi, 416. see also typhoid<br />

Saudi Arabia. see also Middle East, pilgrimage<br />

to Mecca (Hajj and Umra), 116, 120, 129, 290<br />

school teachers, vaccination recommendations,<br />

170<br />

school-based vaccination programs, 28<br />

screening. see pre-vaccination screening<br />

seizures, persons with family history, and<br />

measles vaccination, 278<br />

septic arthritis, 191<br />

septicaemia, 283. see also meningococcal disease<br />

serological testing for immunity, 41<br />

hepatitis A, 206<br />

hepatitis B, 225<br />

measles, 274–275<br />

mumps, 300<br />

rabies and other lyssaviruses (including<br />

<strong>Australian</strong> bat lyssavirus), 369<br />

rubella, 390–391, 395<br />

varicella, 429<br />

zoster (herpes zoster), 452–453<br />

serum, in vaccines, 469, 486<br />

severe combined immunodeficiency, 16, 379<br />

severe dehydrating gastroenteritis, 372. see also<br />

rotavirus<br />

sex industry workers, 11, 175<br />

hepatitis A, 204, 205<br />

hepatitis B, 223, 224, 225<br />

shearers. see animals, persons working with<br />

shingles. see zoster (herpes zoster)<br />

Shingles Prevention Study, 447–448<br />

short gut syndrome, and rotavirus vaccination,<br />

380<br />

sickle cell disease, and pneumococcal disease,<br />

326–327<br />

INDEX 519<br />

INDEX


simultaneous injections, 9, 84<br />

sinusitis, 317<br />

skin cleaning for vaccination, 70<br />

skin disease, and tuberculosis vaccination, 414<br />

skin penetration procedures, vaccination<br />

recommendations, 172, 209, 210, 223<br />

skin test. see Q-Vax Skin Test; tuberculin skin<br />

test<br />

smallpox, 1, 7, 104, 171, 482<br />

smokers. see tobacco smoking<br />

sodium borate, in vaccines, 470<br />

solid organ transplant (SOT) recipients<br />

hepatitis A, 204<br />

hepatitis B, 222<br />

rotavirus, 373<br />

vaccination recommendations, 150–154,<br />

151–154<br />

sorbitol, in vaccines, 19, 472, 485<br />

South Australia<br />

adverse events following immunisation<br />

(AEFI) notification, 95, 96<br />

age of consent, 26<br />

<strong>Australian</strong> Red Cross Blood Service, 457<br />

government health authority and<br />

communicable disease control contact<br />

details, 465–466<br />

hepatitis A, 105, 107, 199, 203, 204<br />

online ‘catch-up calculator,’ 39<br />

pneumococcal disease, 52, 57, 57–58, 105,<br />

108, 318, 324<br />

tuberculosis, 106, 408<br />

special risk groups, 9–11, 104–175. see also under<br />

risk group names<br />

Aboriginal and Torres Strait Islander<br />

people, 35, 104–112<br />

adverse event following immunisation,<br />

130–133<br />

anaesthesia or surgery, 36, 168<br />

bleeding disorders, 168<br />

blood product recipients, 33, 166–167<br />

correctional facility inmates, 174<br />

drugs, persons who inject, 175<br />

men who have sex with men, 174<br />

migrants to Australia, 173–174<br />

normal human immunoglobulin (NHIG)<br />

recipients, 166–167<br />

occupational risks, 169–173<br />

sex industry workers, 175<br />

travellers, 127–128<br />

spica casts, injection sites, 74<br />

splenectomy. see asplenia, persons with<br />

Stamaril, 440<br />

adverse events following immunisation<br />

(AEFI), 444<br />

dosage and administration, 126<br />

and egg protein, 132<br />

520 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

lower age limits for, 128<br />

transport, storage and handling, 440<br />

status of vaccination. see vaccination status and<br />

history<br />

steroids, and zoster vaccination, 453<br />

straddle position, 77–78<br />

Streptococcus pneumoniae, 108, 155, 317. see also<br />

pneumococcal disease<br />

subcutaneous (SC) injection, 68–69, 72, 82<br />

sucrose, in vaccines, 19, 485<br />

sudden infant death syndrome (SIDS), 94, 143<br />

and pertussis vaccination, 313<br />

surgery, vaccination recommendations before<br />

or after, 168<br />

Synagis, 457, 462<br />

Synflorix, 320<br />

dosage and administration, 194, 323<br />

systemic lupus erythematosus, vaccination<br />

recommendations, 165<br />

T<br />

Tasmania<br />

adverse events following immunisation<br />

(AEFI) notification, 96<br />

age of consent, 26<br />

<strong>Australian</strong> Red Cross Blood Service, 457<br />

government health authority and<br />

communicable disease control contact<br />

details, 465–466<br />

pneumococcal disease, 52, 56<br />

tattooists. see skin penetration procedures<br />

tetanus, 397–407<br />

adverse events following immunisation<br />

(AEFI), 405<br />

bacteriology, 397<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 11–12<br />

clinical features, 397<br />

contraindications to vaccination, 404<br />

dosage and administration, 401<br />

epidemiology, 398<br />

precautions, 404–405<br />

pregnancy and breastfeeding, 402<br />

public health management, 405<br />

recommendations, 401–402<br />

tetanus-prone wounds, 402–404<br />

transport, storage and handling, 401<br />

and travellers, 117<br />

vaccines, 398–400<br />

variations from product information,<br />

405–407<br />

tetanus immunoglobulin (TIG), 403–404<br />

availability, 457<br />

intramuscular use, 462–463<br />

intravenous use, 463<br />

Tetanus Immunoglobulin-VF, 405, 463. see also<br />

tetanus immunoglobulin (TIG)


<strong>The</strong>rapeutic Goods Administration (TGA), 5, 7,<br />

18, 94, 461, 482, 486<br />

adverse events following immunisation<br />

(AEFI) notification, 96, 134<br />

direct reporting to, 96–97<br />

vaccine components, 469–472<br />

thimerosal. see thiomersal, in vaccines<br />

thiomersal, in vaccines, 19, 347, 469, 484<br />

thrombocytopenia, 493<br />

and measles vaccination, 278, 279<br />

and passive immunisation, 464<br />

and rubella vaccination, 384, 394<br />

and varicella vaccination, 423, 434<br />

thymus disorders, and yellow fever<br />

vaccination, 444<br />

tick-borne encephalitis, and travellers, 121<br />

tobacco smoking, and pneumococcal disease,<br />

110, 327, 331<br />

Torres Strait Islanders. see Aboriginal and<br />

Torres Strait Islander people<br />

Torres Strait Islands. see also Queensland, and<br />

Japanese encephalitis vaccination, 111, 172,<br />

264<br />

transplants. see haematopoietic stem cell<br />

transplant (HSCT) recipients; solid organ<br />

transplant (SOT) recipients<br />

transport, storage and handling of vaccines, 7,<br />

24–25. see also under individual disease names<br />

travellers. see also Africa; Americas; Asia;<br />

Europe; Middle East; Oceania; Saudi Arabia<br />

children – lower age limits for vaccines,<br />

127–128<br />

cholera, 119<br />

common infections, 113–114<br />

diphtheria, 117<br />

dosage and administration, 123–126<br />

hepatitis A, 119, 204<br />

hepatitis B, 117–118, 224<br />

immunocompromised persons, 147<br />

influenza, 118, 256<br />

Japanese encephalitis, 119, 263–264<br />

measles, 274<br />

meningococcal disease, 120, 291<br />

MMRV (measles-mumps-rubella-varicella)<br />

vaccine, 118<br />

online information sources on health risks<br />

for, 128–129<br />

pertussis, 117<br />

pilgrimage to Mecca (Hajj and Umra), 16,<br />

120, 129, 290<br />

pneumococcal disease, 118<br />

poliomyelitis, 118, 342<br />

pre-travel information to acquire, 114–117<br />

rabies, 120, 358<br />

routinely recommended vaccines, 117–118<br />

special risk groups, 127–128<br />

tetanus, 117<br />

tick-borne encephalitis vaccination, 121<br />

tuberculosis, 121<br />

typhoid, 122, 417, 419–420<br />

vaccination recommendations, 36<br />

vaccine documentation, 116–117<br />

vaccines based on itinerary, 119–126<br />

vaccines required by International Health<br />

Regulations, 116<br />

yellow fever, 122, 440, 441–442<br />

Tripacel, 184, 306, 400<br />

variations from product information, 189,<br />

315, 406<br />

tuberculin skin test, 116, 121, 410, 413<br />

and measles-mumps-rubella (MMR)<br />

vaccination, 278<br />

tuberculosis (TB), 408–415<br />

adverse events following immunisation<br />

(AEFI), 414<br />

bacteriology, 408<br />

BCG vaccination procedures, 411<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 16<br />

clinical features, 408<br />

contraindications to vaccination, 413<br />

dosage and administration, 410–411<br />

epidemiology, 408–409<br />

and Indigenous neonates, 106, 412<br />

precautions, 121, 414<br />

pregnancy and breastfeeding, 413<br />

public health management, 414<br />

recommendations, 412–413<br />

transport, storage and handling, 410<br />

and travellers, 121<br />

variations from product information, 414<br />

Twinrix (720/20), 200, 212<br />

dosage and administration, 125, 202, 217<br />

lower age limits for, 202<br />

vaccination recommendations, 205<br />

vaccination schedules, 215, 217<br />

vaccination schedules, accelerated, 216<br />

Twinrix Junior (360/10), 200, 211–212<br />

dosage and administration, 202, 217<br />

lower age limits for, 127<br />

recommended doses and schedules, 202<br />

vaccination schedule, 215<br />

Typherix, 417<br />

dosage and administration, 126, 419<br />

lower age limits for, 128<br />

Typhim Vi, 417<br />

dosage and administration, 126, 419<br />

lower age limits for, 128<br />

variations from product information, 422<br />

typhoid, 416–422<br />

bacteriology, 416<br />

clinical features, 416<br />

co-administration with other vaccines, 419<br />

contraindications to vaccination, 421<br />

INDEX 521<br />

INDEX


dosage and administration, 418–419<br />

epidemiology, 416–417<br />

and HIV-infected persons, 159<br />

precautions, 421–422<br />

pregnancy and breastfeeding, 137, 139, 421<br />

public health management, 422<br />

recommendations, 419–420<br />

route of vaccine administration, 68–69<br />

transport, storage and handling, 418<br />

and travellers, 122<br />

vaccines, 417–418<br />

variations from product information, 422<br />

V<br />

vaccination, commonly asked questions,<br />

473–488<br />

vaccination contraindications. see<br />

contraindications to vaccination<br />

vaccination documentation. see documentation<br />

of vaccination<br />

vaccination procedures. see procedures for<br />

vaccination<br />

vaccination recommendations. see under<br />

individual disease names<br />

vaccination status and history, 39–40<br />

vaccine administration. see administration of<br />

vaccines<br />

vaccine availability in Australia, 498–499<br />

vaccine components, commonly asked<br />

questions about, 484–486<br />

vaccine effectiveness, 20–21<br />

vaccine efficacy, 20–21<br />

vaccine failure, 20–21<br />

Vaccine Information and Vaccine<br />

Administration System (VIVAS), 102–103<br />

vaccine preparation. see preparation of vaccines<br />

vaccine product information, 5, 6, 19. see also<br />

under individual disease names, variations from<br />

product information<br />

vaccine safety, 21–23. see also adverse events<br />

following immunisation (AEFI)<br />

commonly asked questions about, 481–483<br />

vaccine side effects. see adverse events<br />

following immunisation (AEFI)<br />

vaccine-preventable diseases, 175–455. see also<br />

under individual disease names<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 11–16<br />

cholera, 11, 176–181<br />

diphtheria, 11–12, 182–190<br />

Haemophilus influenzae type b (Hib), 12,<br />

191–197<br />

hepatitis A, 12, 198–207<br />

hepatitis B, 12, 208–230<br />

human papillomavirus (HPV), 13, 231–242<br />

influenza, 13, 243–258<br />

Japanese encephalitis, 13, 259–266<br />

522 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition<br />

measles, 13–14, 267–282<br />

meningococcal disease, 14, 283–294<br />

mumps, 13–14, 295–301<br />

pertussis, 11–12, 302–316<br />

pneumococcal disease, 14–15, 317–337<br />

poliomyelitis, 15, 338–344<br />

Q fever, 15, 345–352<br />

rabies and other lyssaviruses (including<br />

<strong>Australian</strong> bat lyssavirus), 15–16, 353–371<br />

rotavirus, 16, 372–383<br />

rubella, 13–14, 384–396<br />

tetanus, 11–12, 397–407<br />

tuberculosis, 16, 408–415<br />

typhoid, 416–422<br />

varicella, 13–14, 423–438<br />

yellow fever, 16, 439–445<br />

zoster (herpes zoster), 16, 446–455<br />

vaginal cancer, 233<br />

valid consent. see consent<br />

valid dose. see dosage<br />

Vaqta Adult formulation, 200<br />

dosage and administration, 125, 202<br />

vaccine components, 469–472<br />

Vaqta Paediatric/Adolescent formulation, 200<br />

dosage and administration, 202<br />

lower age limits for, 127<br />

vaccine components, 469–472<br />

varicella, 423–438<br />

adverse events following immunisation<br />

(AEFI), 433–435<br />

catch-up vaccination, 52, 63–64<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 13–14<br />

and children, 427–428<br />

clinical features, 423<br />

co-administration with other vaccines, 427<br />

contraindications to vaccination, 431–432<br />

dosage and administration, 49, 50, 427<br />

epidemiology, 424<br />

higher risk groups, 430<br />

and HIV-infected persons, 159<br />

interchangeability of vaccines, 427<br />

minimum acceptable age for the 1st vaccine<br />

dose, 47<br />

post-exposure vaccination, 429–430, 437<br />

precautions, 432–433<br />

pregnancy and breastfeeding, 140, 430–431,<br />

432<br />

public health management, 435–437<br />

recommendations, 427–430<br />

serological testing for immunity, 429<br />

transport, storage and handling, 426–427<br />

and travellers, 118<br />

vaccines, 68–69, 424–426<br />

variations from product information,<br />

437–438<br />

virology, 423


varicella-zoster immunoglobulin, 457<br />

varicella-zoster virus (VZV), 423, 446. see also<br />

varicella; zoster (herpes zoster)<br />

Varilrix, 48, 425<br />

dosage and administration, 124<br />

transport, storage and handling, 426<br />

vaccine components, 469, 469–472, 471<br />

variations from product information, 437<br />

Varivax Refrigerated, 425<br />

dosage and administration, 124<br />

transport, storage and handling, 427<br />

vaccine components, 469, 471<br />

variations from product information, 437,<br />

438<br />

vasovagal episode, 87–88<br />

vastus lateralis injection site, 73, 75, 78, 79–80,<br />

474<br />

Vaxigrip, 247<br />

vaccine components, 470–471<br />

Vaxigrip Junior, 247<br />

ventrogluteal injection site, 73, 74, 76, 78, 80–81,<br />

83, 84, 474<br />

veterinarians. see animals, persons working<br />

with<br />

Vibrio cholerae, 119, 176. see also cholera<br />

Victoria<br />

adverse events following immunisation<br />

(AEFI) notification, 95, 96<br />

age of consent, 26<br />

<strong>Australian</strong> Red Cross Blood Service, 457<br />

government health authority and<br />

communicable disease control contact<br />

details, 465–466<br />

pneumococcal disease, 52, 56, 322<br />

pneumococcal vaccination, 52, 56<br />

Victorian Cytology Service, 101<br />

viral haemorrhagic fever. see yellow fever<br />

Vivaxim, 200, 417<br />

dosage and administration, 125–126, 202,<br />

419<br />

recommended doses and schedules, 202<br />

Vivotif Oral, 417–418<br />

dosage and administration, 126<br />

lower age limits for, 128<br />

variations from product information, 422<br />

vulval cancer, 233<br />

W<br />

Western Australia<br />

adverse events following immunisation<br />

(AEFI) notification, 95, 96<br />

age of consent, 26<br />

<strong>Australian</strong> Red Cross Blood Service, 457<br />

government health authority and<br />

communicable disease control contact<br />

details, 465–466<br />

hepatitis A, 105, 107, 199, 203, 204<br />

hepatitis A vaccine recommendations for<br />

Indigenous persons, 203<br />

influenza, 92<br />

pneumococcal disease, 15, 52, 57, 105, 108,<br />

318, 324<br />

pneumococcal vaccination, 57–58<br />

tuberculosis, 106, 107<br />

whooping cough. see pertussis<br />

wildlife workers. see animals, persons working<br />

with<br />

women. see also breastfeeding; human<br />

papillomavirus, female vaccination;<br />

pregnancy<br />

planning pregnancy, pregnant or<br />

breastfeeding, 133–142<br />

rubella vaccination for women of childbaring<br />

age, 390, 391<br />

World Health Organization Global Advisory<br />

Committee on Vaccine Safety, 228<br />

World Health Organization (WHO)<br />

global elimination of measles, 268<br />

Global Polio Eradication Initiative, 338<br />

immunisation schedules for other<br />

countries, 173<br />

influenza pandemic, 245–246<br />

rabies vaccine recommendations, 358, 363,<br />

367<br />

and rubella vaccination, 385<br />

serological testing for immunity to rubella,<br />

391<br />

and tuberculosis, 408<br />

yellow fever vaccination requirements, 442<br />

wounds<br />

management after rabies or lyssavirus<br />

exposure, 357, 361, 366, 367<br />

tetanus prophylaxis for, 403–404<br />

tetanus-prone wounds, 402–404<br />

Y<br />

yeast, in vaccines, 472<br />

yellow fever, 68–69<br />

adverse events following immunisation<br />

(AEFI), 444<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 16<br />

clinical features, 439<br />

co-administration with other vaccines, 441<br />

contraindications to vaccination, 443–444<br />

dosage and administration, 440–441<br />

and egg allergies, 132–133<br />

epidemiology, 439–440<br />

and HIV-infected persons, 159<br />

and immunocompromised persons, 148<br />

precautions, 444<br />

pregnancy and breastfeeding, 139, 443, 445<br />

public health management, 445<br />

recommendations, 441–442<br />

INDEX 523<br />

INDEX


transport, storage and handling, 440<br />

and travellers, 122<br />

vaccine, 440<br />

variations from product information, 445<br />

virology, 439<br />

yellow fever vaccine-associated viscerotropic<br />

disease, 444, 445<br />

Z<br />

Zagreb, 363<br />

zoo workers. see animals, persons working<br />

with<br />

Zostavax, 323–324, 447–449<br />

adverse events following immunisation<br />

(AEFI), 454<br />

dosage and administration, 449–450<br />

precautions, 454<br />

transport, storage and handling, 449<br />

vaccination recommendations, 452<br />

variations from product information, 337,<br />

455<br />

zoster (herpes zoster), 446–455<br />

adverse events following immunisation<br />

(AEFI), 454<br />

catch-up vaccination, 63–64<br />

changes in the <strong>10th</strong> edition of the<br />

handbook, 16<br />

clinical features, 446<br />

co-administration with other vaccines,<br />

449–450, 453<br />

contraindications to vaccination, 453<br />

dosage and administration, 449–450<br />

epidemiology, 446–447<br />

precautions, 453–454<br />

pregnancy and breastfeeding, 141, 453<br />

and recipients of normal human<br />

immunoglobulin and other blood<br />

products, 166<br />

recommendations, 147, 159, 450–453<br />

serological testing for immunity, 452–453<br />

transport, storage and handling, 449<br />

vaccine, 68–69, 447–449<br />

variations from product information, 454<br />

virology, 446<br />

zoster immunoglobulin (ZIG), 460. see also<br />

Zoster Immunoglobulin-VF (human)<br />

Zoster Immunoglobulin-VF (human), 435–437<br />

variations from product information, 438<br />

524 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition


INDEX 525<br />

INDEX


526 <strong>The</strong> <strong>Australian</strong> <strong>Immunisation</strong> <strong>Handbook</strong> <strong>10th</strong> edition

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!